Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38903085

ABSTRACT

The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine F-actin structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of Mrtf, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G-protein-coupled receptor (GPCR) Smog, G-protein αq subunit, Rho1 GTPase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand Fog to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, a NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.

2.
Curr Biol ; 33(22): R1205-R1207, 2023 11 20.
Article in English | MEDLINE | ID: mdl-37989102

ABSTRACT

A new study shows that cell size, in conjunction with specific signaling pathways, controls apoptosis within developing tissues. Cells with smaller sizes and relatively smaller sizes compared to their neighbors exhibit an increased likelihood of undergoing apoptosis. These processes are regulated by the Hippo/YAP and Notch pathways, respectively.


Subject(s)
Protein Serine-Threonine Kinases , Signal Transduction , Protein Serine-Threonine Kinases/metabolism , Cell Differentiation , Cell Proliferation , Homeostasis , Cell Size
3.
Dev Cell ; 58(19): 1933-1949.e5, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37567172

ABSTRACT

The ability of stem cells to switch between quiescent and proliferative states is crucial for maintaining tissue homeostasis and regeneration. In Drosophila, quiescent neural stem cells (qNSCs) extend a primary protrusion, a hallmark of qNSCs. Here, we have found that qNSC protrusions can be regenerated upon injury. This regeneration process relies on the Golgi apparatus that acts as the major acentrosomal microtubule-organizing center in qNSCs. A Golgi-resident GTPase Arf1 and its guanine nucleotide exchange factor Sec71 promote NSC reactivation and regeneration via the regulation of microtubule growth. Arf1 physically associates with its new effector mini spindles (Msps)/XMAP215, a microtubule polymerase. Finally, Arf1 functions upstream of Msps to target the cell adhesion molecule E-cadherin to NSC-neuropil contact sites during NSC reactivation. Our findings have established Drosophila qNSCs as a regeneration model and identified Arf1/Sec71-Msps pathway in the regulation of microtubule growth and NSC reactivation.

4.
EMBO Rep ; 24(9): e56624, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37440685

ABSTRACT

The ability of stem cells to switch between quiescent and proliferative states is crucial for maintaining tissue homeostasis and regeneration. Drosophila quiescent neural stem cells (qNSCs) extend a primary protrusion that is enriched in acentrosomal microtubules and can be regenerated upon injury. Arf1 promotes microtubule growth, reactivation (exit from quiescence), and regeneration of qNSC protrusions upon injury. However, how Arf1 is regulated in qNSCs remains elusive. Here, we show that the microtubule minus-end binding protein Patronin/CAMSAP promotes acentrosomal microtubule growth and quiescent NSC reactivation. Patronin is important for the localization of Arf1 at Golgi and physically associates with Arf1, preferentially with its GDP-bound form. Patronin is also required for the regeneration of qNSC protrusion, likely via the regulation of microtubule growth. Finally, Patronin functions upstream of Arf1 and its effector Msps/XMAP215 to target the cell adhesion molecule E-cadherin to NSC-neuropil contact sites during NSC reactivation. Our findings reveal a novel link between Patronin/CAMSAP and Arf1 in the regulation of microtubule growth and NSC reactivation. A similar mechanism might apply to various microtubule-dependent systems in mammals.


Subject(s)
Drosophila Proteins , Neural Stem Cells , Animals , Microtubule-Associated Proteins/metabolism , Drosophila/metabolism , Microtubules/metabolism , Drosophila Proteins/metabolism , Neural Stem Cells/metabolism , Mammals/metabolism
5.
Dev Cell ; 58(4): 267-277.e5, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36800994

ABSTRACT

The number of cells in tissues is controlled by cell division and cell death, and its misregulation could lead to pathological conditions such as cancer. To maintain the cell numbers, a cell-elimination process called apoptosis also stimulates the proliferation of neighboring cells. This mechanism, apoptosis-induced compensatory proliferation, was originally described more than 40 years ago. Although only a limited number of the neighboring cells need to divide to compensate for the apoptotic cell loss, the mechanisms that select cells to divide have remained elusive. Here, we found that spatial inhomogeneity in Yes-associated protein (YAP)-mediated mechanotransduction in neighboring tissues determines the inhomogeneity of compensatory proliferation in Madin-Darby canine kidney (MDCK) cells. Such inhomogeneity arises from the non-uniform distribution of nuclear size and the non-uniform pattern of mechanical force applied to neighboring cells. Our findings from a mechanical perspective provide additional insight into how tissues precisely maintain homeostasis.


Subject(s)
Apoptosis , Mechanotransduction, Cellular , Animals , Dogs , Apoptosis/physiology , Cell Death , Cell Division , Madin Darby Canine Kidney Cells , Cell Proliferation/physiology
6.
Nat Phys ; 19: 132-141, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36686215

ABSTRACT

Epithelia act as a barrier against environmental stress and abrasion and in vivo they are continuously exposed to environments of various mechanical properties. The impact of this environment on epithelial integrity remains elusive. By culturing epithelial cells on 2D hydrogels, we observe a loss of epithelial monolayer integrity through spontaneous hole formation when grown on soft substrates. Substrate stiffness triggers an unanticipated mechanical switch of epithelial monolayers from tensile on soft to compressive on stiff substrates. Through active nematic modelling, we find that spontaneous half-integer defect formation underpinning large isotropic stress fluctuations initiate hole opening events. Our data show that monolayer rupture due to high tensile stress is promoted by the weakening of cell-cell junctions that could be induced by cell division events or local cellular stretching. Our results show that substrate stiffness provides feedback on monolayer mechanical state and that topological defects can trigger stochastic mechanical failure, with potential application towards a mechanistic understanding of compromised epithelial integrity during immune response and morphogenesis.

7.
Biomech Model Mechanobiol ; 21(5): 1511-1530, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36057053

ABSTRACT

Tissue layers can generally slide at the interface, accompanied by the dissipation due to friction. Nevertheless, it remains elusive how force could propagate in a tissue with such interfacial friction. Here, we elaborate the force dynamics in a prototypical multilayer system in which an epithelial monolayer was cultivated upon an elastic substrate in contact with a hard surface, and discover a novel mechanism of pronounced force propagation over a long distance due to interfacial dynamics between substrate layers. We derived an analytical model for the dynamics of the elastic substrate under the shear stress provided by the cell layer at the surface boundary and the friction at bottom. The model reveals that sliding between substrate layers leads to an expanding stretch regime from a shear regime of substrate deformation in time and space. The regime boundary propagating diffusively with a speed depending on the stiffness, thickness, and slipperiness of the substrate, is a robust nature of a deformed elastic sheet with interfacial friction. These results shed new light on force propagation in tissues and our model could serve as a basis for studies of such propagation in a more complex tissue environment.


Subject(s)
Mechanical Phenomena , Friction , Elasticity , Stress, Mechanical
8.
Front Cell Dev Biol ; 9: 669086, 2021.
Article in English | MEDLINE | ID: mdl-34222239

ABSTRACT

Cadherin-mediated adhesions (also known as adherens junctions) are adhesive complexes that connect neighboring cells in a tissue. While the role of the actin cytoskeleton in withstanding tension at these sites of contact is well documented, little is known about the involvement of microtubules and the associated endoplasmic reticulum (ER) network in cadherin mechanotransduction. Therefore, we investigated how the organization of ER extensions in close proximity of cadherin-mediated adhesions can affect such complexes, and vice versa. Here, we show that the extension of the ER to cadherin-mediated adhesions is tension dependent and appears to be cadherin-type specific. Furthermore, the different structural organization of the ER/microtubule network seems to affect the localization of ER-bound PTP1B at cadherin-mediated adhesions. This phosphatase is involved in the modulation of vinculin, a molecular clutch which enables differential engagement of the cadherin-catenin layer with the actomyosin cytoskeleton in response to tension. This suggests a link between structural organization of the ER/microtubule network around cadherin-specific adhesions, to control the mechanotransduction of adherens junctions by modulation of vinculin conformational state.

9.
New Phytol ; 232(2): 928-940, 2021 10.
Article in English | MEDLINE | ID: mdl-34270808

ABSTRACT

The evolution of herbicide resistance in weeds is an example of parallel evolution, through which genes encoding herbicide target proteins are repeatedly represented as evolutionary targets. The number of herbicide target-site genes differs among species, and little is known regarding the effects of duplicate gene copies on the evolution of herbicide resistance. We investigated the evolution of herbicide resistance in Monochoria vaginalis, which carries five copies of sulfonylurea target-site acetolactate synthase (ALS) genes. Suspected resistant populations collected across Japan were investigated for herbicide sensitivity and ALS gene sequences, followed by functional characterization and ALS gene expression analysis. We identified over 60 resistant populations, all of which carried resistance-conferring amino acid substitutions exclusively in MvALS1 or MvALS3. All MvALS4 alleles carried a loss-of-function mutation. Although the enzymatic properties of ALS encoded by these genes were not markedly different, the expression of MvALS1 and MvALS3 was prominently higher among all ALS genes. The higher expression of MvALS1 and MvALS3 is the driving force of the biased representation of genes during the evolution of herbicide resistance in M. vaginalis. Our findings highlight that gene expression is a key factor in creating evolutionary hotspots.


Subject(s)
Acetolactate Synthase , Herbicides , Acetolactate Synthase/genetics , Gene Expression , Herbicide Resistance/genetics , Herbicides/pharmacology , Plant Proteins/genetics , Plant Weeds/genetics
10.
Semin Cell Dev Biol ; 120: 171-180, 2021 12.
Article in English | MEDLINE | ID: mdl-34167884

ABSTRACT

Adult epidermal development in Drosophila showcases a striking balance between en masse spreading of the developing adult precursor tissues and retraction of the degenerating larval epidermis. The adult precursor tissues, driven by both intrinsic plasticity and extrinsic mechanical cues, shape the segments of the adult epidermis and appendages. Here, we review the tissue architectural changes that occur during epidermal morphogenesis in the Drosophila pupa, with a particular emphasis on the underlying mechanical principles. We highlight recent developments in our understanding of adult epidermal morphogenesis. We further discuss the forces that drive these morphogenetic events and finally outline open questions and challenges.


Subject(s)
Epidermal Cells/metabolism , Morphogenesis/physiology , Animals , Drosophila , Pupa
11.
ACS Biomater Sci Eng ; 7(6): 2661-2675, 2021 06 14.
Article in English | MEDLINE | ID: mdl-33942605

ABSTRACT

Endothelial Cells (ECs) form cohesive cellular lining of the vasculature and play essential roles in both developmental processes and pathological conditions. Collective migration and proliferation of endothelial cells (ECs) are key processes underlying endothelialization of vessels as well as vascular graft, but the complex interplay of mechanical and biochemical signals regulating these processes are still not fully elucidated. While surface topography and biochemical modifications have been used to enhance endothelialization in vitro, thus far such single-modality modifications have met with limited success. As combination therapy that utilizes multiple modalities has shown improvement in addressing various intractable and complex biomedical conditions, here, we explore a combined strategy that utilizes topographical features in conjunction with pharmacological perturbations. We characterized EC behaviors in response to micrometer-scale grating topography in concert with pharmacological perturbations of endothelial adherens junctions (EAJ) regulators. We found that the protein tyrosine phosphatase, PTP1B, serves as a potent regulator of EAJ stability, with PTP1B inhibition synergizing with grating topographies to modulate EAJ rearrangement, thereby augmenting global EC monolayer sheet orientation, proliferation, connectivity, and collective cell migration. Our data delineates the crosstalk between cell-ECM topography sensing and cell-cell junction integrity maintenance and suggests that the combined use of grating topography and PTP1B inhibitor could be a promising strategy for promoting collective EC migration and proliferation.


Subject(s)
Adherens Junctions , Endothelial Cells , Cell Line , Cell Movement , Endothelium, Vascular
13.
Adv Biol (Weinh) ; 5(6): e2000493, 2021 06.
Article in English | MEDLINE | ID: mdl-33788418

ABSTRACT

Accurate segregation of chromosomes during anaphase relies on the central spindle and its regulators. A newly raised concept of the central spindle, the bridging fiber, shows that sliding of antiparallel microtubules (MTs) within the bridging fiber promotes chromosome segregation. However, the regulators of the bridging fiber and its regulatory mechanism on MTs sliding remain largely unknown. In this study, the non-motor microtubule-associated protein, hyaluronan-mediated motility receptor (HMMR), is identified as a novel regulator of the bridging fiber. It then identifies that HMMR regulates MTs sliding within the bridging fiber by cooperating with its binding partner HSET. By utilizing a laser-based cell ablation system and photoactivation approach, the study's results reveal that depletion of HMMR causes an inhibitory effect on MTs sliding within the bridging fiber and disrupts the forced uniformity on the kinetochore-attached microtubules-formed fibers (k-fibers). These are created by suppressing the dynamics of HSET, which functions in transiting the force from sliding of bridging MTs to the k-fiber. This study sheds new light on the novel regulatory mechanism of MTs sliding within the bridging fiber by HMMR and HSET and uncovers the role of HMMR in chromosome segregation during anaphase.


Subject(s)
Chromosome Segregation , Spindle Apparatus , Anaphase , Extracellular Matrix Proteins , Hyaluronan Receptors , Microtubules
14.
Nat Mater ; 20(8): 1156-1166, 2021 08.
Article in English | MEDLINE | ID: mdl-33603188

ABSTRACT

Actomyosin machinery endows cells with contractility at a single-cell level. However, within a monolayer, cells can be contractile or extensile based on the direction of pushing or pulling forces exerted by their neighbours or on the substrate. It has been shown that a monolayer of fibroblasts behaves as a contractile system while epithelial or neural progentior monolayers behave as an extensile system. Through a combination of cell culture experiments and in silico modelling, we reveal the mechanism behind this switch in extensile to contractile as the weakening of intercellular contacts. This switch promotes the build-up of tension at the cell-substrate interface through an increase in actin stress fibres and traction forces. This is accompanied by mechanotransductive changes in vinculin and YAP activation. We further show that contractile and extensile differences in cell activity sort cells in mixtures, uncovering a generic mechanism for pattern formation during cell competition, and morphogenesis.


Subject(s)
Actomyosin/metabolism , Mechanical Phenomena , Biomechanical Phenomena , Cell Movement , Computer Simulation , Models, Biological
15.
Nat Commun ; 12(1): 397, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452264

ABSTRACT

Apoptotic extrusion is crucial in maintaining epithelial homeostasis. Current literature supports that epithelia respond to extrusion by forming a supracellular actomyosin purse-string in the neighbors. However, whether other actin structures could contribute to extrusion and how forces generated by these structures can be integrated are unknown. Here, we found that during extrusion, a heterogeneous actin network composed of lamellipodia protrusions and discontinuous actomyosin cables, was reorganized in the neighboring cells. The early presence of basal lamellipodia protrusion participated in both basal sealing of the extrusion site and orienting the actomyosin purse-string. The co-existence of these two mechanisms is determined by the interplay between the cell-cell and cell-substrate adhesions. A theoretical model integrates these cellular mechanosensitive components to explain why a dual-mode mechanism, which combines lamellipodia protrusion and purse-string contractility, leads to more efficient extrusion than a single-mode mechanism. In this work, we provide mechanistic insight into extrusion, an essential epithelial homeostasis process.


Subject(s)
Actomyosin/metabolism , Apoptosis/physiology , Cell Adhesion/physiology , Epithelium/physiology , Models, Biological , Animals , Dogs , Madin Darby Canine Kidney Cells , Pseudopodia/physiology
16.
Curr Opin Cell Biol ; 67: 141-146, 2020 12.
Article in English | MEDLINE | ID: mdl-33189987

ABSTRACT

Organism development requires fine-tuning of the cell number by apoptosis and cell division, as well as proper cell fate specification. These processes are achieved through the integration of intracellular signals and intercellular interactions with neighboring cells as well as the extracellular environment. Apoptosis, a form of cell death typically associated with development and homeostasis, is mainly regulated by the caspase family of proteases. Although caspases are known to initiate and execute apoptosis, it is also known that low caspase levels have a broad spectrum of nonapoptotic functions, including differentiation and organ growth. These different roles of caspases raise intriguing questions: how are caspase levels regulated and what defines the balance between life and death? In this review, we focus on some recent findings that highlight how nonlethal levels of caspase activity, transcriptional coregulator Yes-associated protein (YAP), and mechanical factors influence each other in determining cell fate. We further discuss a possibility that the mechanical signals encountered by cells could regulate the level of caspase activity by mechanics through YAP and, in turn, how this determines whether a cell is susceptible or resistant to undergoing apoptosis in response to cell death stimuli.


Subject(s)
Apoptosis , Caspases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Biomechanical Phenomena , Cell Differentiation , Cell Lineage , Models, Biological
17.
Curr Biol ; 30(17): 3364-3377.e4, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32679105

ABSTRACT

Actomyosin networks provide the major contractile machinery for regulating cell and tissue morphogenesis during development. These networks undergo dynamic rearrangements, enabling cells to have a broad range of mechanical actions. How cells integrate different mechanical stimuli to accomplish complicated tasks in vivo remains unclear. Here, we explore this problem in the context of cell matching, where individual cells form precise inter-cellular connections between partner cells. To study the dynamic roles of actomyosin networks in regulating precise cell matching, we focused on the process of heart formation during Drosophila embryogenesis, where selective filopodia-binding adhesions ensure precise cell alignment. We find that non-muscle Myosin II clusters periodically oscillate within cardioblasts with ~4-min intervals. We observe that filopodia dynamics-including protrusions, retraction, binding stabilization, and binding separation-are correlated with the periodic localization of Myosin II clusters at the cell leading edge. Perturbing the Myosin II activity and oscillatory pattern alters the filopodia properties and binding dynamics and results in mismatched cardioblasts. By simultaneously changing the activity of Myosin II and filopodia adhesion levels, we further demonstrate that levels of Myosin II and adhesion are balanced to ensure precise connectivity between cardioblasts. Combined, we propose a mechanical proofreading machinery of robust cell matching, whereby oscillations of Myosin II within cardioblasts periodically probe filopodia adhesion strength and ensure correct cell-cell connection formation.


Subject(s)
Cell Movement , Drosophila Proteins/metabolism , Drosophila/embryology , Heart/embryology , Morphogenesis , Myosin Type II/metabolism , Pseudopodia/physiology , Actomyosin/metabolism , Animals , Drosophila/physiology , Drosophila Proteins/genetics , Heart/physiology , Myosin Type II/genetics
18.
Dev Cell ; 53(6): 621-622, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32574589

ABSTRACT

It remains unclear how intercellular force transmission is coupled with intracellular biochemical signaling to achieve collective cell migration. In this issue of Developmental Cell, Hino et al. demonstrate that intracellular ERK signaling propagates as waves across a tissue and couples with intercellular mechano-transduction to direct epithelial cell migration.


Subject(s)
Signal Transduction , Cell Movement
19.
Curr Biol ; 30(4): 682-690.e5, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32004454

ABSTRACT

During apoptosis, or programmed cell death, a dead cell could be expelled from the tissue by coordinated processes between the dying cell and its neighbors. Apoptotic cell extrusion is driven by actomyosin cable formation and its contraction and lamellipodial crawling of the neighboring cells [1-4]. Throughout cell extrusion, the mechanical coupling of epithelia needs to be maintained in order to preserve tissue homeostasis [1]. Although much is known about the regulation of adherens junctions (AJs) in apoptotic cell extrusion [4-7], the role and dynamics of desmosomal junctions (DJs) during this process remain poorly understood. Here, we show that DJs stay intact throughout and are crucial for cell extrusion. Pre-existing DJs between the apoptotic cell and neighboring cells remain intact, even during the formation of de novo DJs between non-dying cells, suggesting the neighboring cells possess two DJs in the middle of apoptotic cell extrusion. We further found that an actomyosin cable formed in the vicinity of DJs upon apoptosis and subsequently deviated from DJs during its constriction. Interestingly, the departure of the actomyosin cable from DJs coincided with the timing when DJs lost their straightness, suggesting a release of junctional tension at DJs and a mechanical coupling between DJs and actomyosin contractility. The depletion of desmoplakin resulted in defective contractility and an inability to form de novo DJs, leading to a failure of apoptotic cell extrusion. Our study provides a framework to explain how desmosomes play pivotal roles in maintaining epithelial sheet integrity during apoptotic cell extrusion.


Subject(s)
Actomyosin/metabolism , Apoptosis/physiology , Cell Transformation, Neoplastic/metabolism , Desmosomes/metabolism , Animals , Dogs , Madin Darby Canine Kidney Cells
20.
Curr Biol ; 30(4): 670-681.e6, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32004455

ABSTRACT

When oncogenic transformation or apoptosis occurs within epithelia, the harmful or dead cells are apically extruded from tissues to maintain epithelial homeostasis. However, the underlying molecular mechanism still remains elusive. In this study, we first show, using mammalian cultured epithelial cells and zebrafish embryos, that prior to apical extrusion of RasV12-transformed cells, calcium wave occurs from the transformed cell and propagates across the surrounding cells. The calcium wave then triggers and facilitates the process of extrusion. IP3 receptor, gap junction, and mechanosensitive calcium channel TRPC1 are involved in calcium wave. Calcium wave induces the polarized movement of the surrounding cells toward the extruding transformed cells. Furthermore, calcium wave facilitates apical extrusion, at least partly, by inducing actin rearrangement in the surrounding cells. Moreover, comparable calcium propagation also promotes apical extrusion of apoptotic cells. Thus, calcium wave is an evolutionarily conserved, general regulatory mechanism of cell extrusion.


Subject(s)
Calcium Signaling/physiology , Cell Transformation, Neoplastic/metabolism , Animals , Dogs , Embryo, Nonmammalian , Madin Darby Canine Kidney Cells , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...