Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.107
Filtrar
1.
Nat Commun ; 15(1): 2477, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38509115

RESUMO

How a developing organ robustly coordinates the cellular mechanics and growth to reach a final size and shape remains poorly understood. Through iterations between experiments and model simulations that include a mechanistic description of interkinetic nuclear migration, we show that the local curvature, height, and nuclear positioning of cells in the Drosophila wing imaginal disc are defined by the concurrent patterning of actomyosin contractility, cell-ECM adhesion, ECM stiffness, and interfacial membrane tension. We show that increasing cell proliferation via different growth-promoting pathways results in two distinct phenotypes. Triggering proliferation through insulin signaling increases basal curvature, but an increase in growth through Dpp signaling and Myc causes tissue flattening. These distinct phenotypic outcomes arise from differences in how each growth pathway regulates the cellular cytoskeleton, including contractility and cell-ECM adhesion. The coupled regulation of proliferation and cytoskeletal regulators is a general strategy to meet the multiple context-dependent criteria defining tissue morphogenesis.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Morfogênese/fisiologia , Citoesqueleto/metabolismo , Transdução de Sinais/fisiologia , Asas de Animais , Drosophila melanogaster/metabolismo
2.
Curr Biol ; 34(4): 683-696.e6, 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38228149

RESUMO

Intricate branching patterns emerge in internal organs due to the recurrent occurrence of simple deformations in epithelial tissues. During murine lung development, epithelial cells in distal tips of the single tube require fibroblast growth factor (FGF) signals emanating from their surrounding mesenchyme to form repetitive tip bifurcations. However, it remains unknown how the cells employ FGF signaling to convert their behaviors to achieve the recursive branching processes. Here, we show a mechano-chemical regulatory system underlying lung branching morphogenesis, orchestrated by extracellular signal-regulated kinase (ERK) as a downstream driver of FGF signaling. We found that tissue-scale curvature regulated ERK activity in the lung epithelium using two-photon live cell imaging and mechanical perturbations. ERK activation occurs specifically in epithelial tissues exhibiting positive curvature, regardless of whether the change in curvature was attributable to morphogenesis or perturbations. Moreover, ERK activation accelerates actin polymerization preferentially at the apical side of cells, mechanically contributing to the extension of the apical membrane, culminating in a reduction of epithelial tissue curvature. These results indicate the existence of a negative feedback loop between tissue curvature and ERK activity that transcends spatial scales. Our mathematical model confirms that this regulatory mechanism is sufficient to generate the recursive branching processes. Taken together, we propose that ERK orchestrates a curvature feedback loop pivotal to the self-organized patterning of tissues.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular , Pulmão , Camundongos , Animais , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Retroalimentação , Fatores de Crescimento de Fibroblastos/metabolismo , Epitélio/metabolismo , Morfogênese/fisiologia , Mesoderma
3.
Dev Cell ; 59(3): 326-338.e5, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38237591

RESUMO

During organ formation, progenitor cells need to acquire different cell identities and organize themselves into distinct structural units. How these processes are coordinated and how tissue architecture(s) is preserved despite the dramatic cell rearrangements occurring in developing organs remain unclear. Here, we identified cellular rearrangements between acinar and ductal progenitors as a mechanism to drive branching morphogenesis in the pancreas while preserving the integrity of the acinar-ductal functional unit. Using ex vivo and in vivo mouse models, we found that pancreatic ductal cells form clefts by protruding and pulling on the acinar basement membrane, which leads to acini splitting. Newly formed acini remain connected to the bifurcated branches generated by ductal cell rearrangement. Insulin growth factor (IGF)/phosphatidylinositol 3-kinase (PI3K) pathway finely regulates this process by controlling pancreatic ductal tissue fluidity, with a simultaneous impact on branching and cell fate acquisition. Together, our results explain how acinar structure multiplication and branch bifurcation are synchronized during pancreas organogenesis.


Assuntos
Fosfatidilinositol 3-Quinase , Fosfatidilinositol 3-Quinases , Camundongos , Animais , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Pâncreas , Células Acinares/metabolismo , Morfogênese/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo
4.
PLoS Biol ; 22(1): e3002093, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38198514

RESUMO

Epithelial branching morphogenesis is an essential process in living organisms, through which organ-specific epithelial shapes are created. Interactions between epithelial cells and their stromal microenvironment instruct branching morphogenesis but remain incompletely understood. Here, we employed fibroblast-organoid or fibroblast-spheroid co-culture systems and time-lapse imaging to reveal that physical contact between fibroblasts and epithelial cells and fibroblast contractility are required to induce mammary epithelial branching. Pharmacological inhibition of ROCK or non-muscle myosin II, or fibroblast-specific knock-out of Myh9 abrogate fibroblast-induced epithelial branching. The process of fibroblast-induced branching requires epithelial proliferation and is associated with distinctive epithelial patterning of yes associated protein (YAP) activity along organoid branches, which is dependent on fibroblast contractility. Moreover, we provide evidence for the in vivo existence of contractile fibroblasts specifically surrounding terminal end buds (TEBs) of pubertal murine mammary glands, advocating for an important role of fibroblast contractility in branching in vivo. Together, we identify fibroblast contractility as a novel stromal factor driving mammary epithelial morphogenesis. Our study contributes to comprehensive understanding of overlapping but divergent employment of mechanically active fibroblasts in developmental versus tumorigenic programs.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Camundongos , Animais , Glândulas Mamárias Animais/metabolismo , Células Epiteliais/metabolismo , Morfogênese/fisiologia , Técnicas de Cocultura , Fibroblastos/metabolismo
5.
Development ; 151(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38165177

RESUMO

Multicellular rosettes are transient epithelial structures that serve as important cellular intermediates in the formation of diverse organs. Using the zebrafish posterior lateral line primordium (pLLP) as a model system, we investigated the role of the RhoA GEF Mcf2lb in rosette morphogenesis. The pLLP is a group of ∼150 cells that migrates along the zebrafish trunk and is organized into epithelial rosettes; these are deposited along the trunk and will differentiate into sensory organs called neuromasts (NMs). Using single-cell RNA-sequencing and whole-mount in situ hybridization, we showed that mcf2lb is expressed in the pLLP during migration. Live imaging and subsequent 3D analysis of mcf2lb mutant pLLP cells showed disrupted apical constriction and subsequent rosette organization. This resulted in an excess number of deposited NMs along the trunk of the zebrafish. Cell polarity markers ZO-1 and Par-3 were apically localized, indicating that pLLP cells are properly polarized. In contrast, RhoA activity, as well as signaling components downstream of RhoA, Rock2a and non-muscle Myosin II, were diminished apically. Thus, Mcf2lb-dependent RhoA activation maintains the integrity of epithelial rosettes.


Assuntos
Sistema da Linha Lateral , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Transdução de Sinais/fisiologia , Movimento Celular/genética , Morfogênese/fisiologia
6.
Dev Biol ; 506: 1-6, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37995916

RESUMO

Tile patterns, in which numerous cells are arranged in a regular pattern, are found in a variety of multicellular organisms and play important functional roles. Such regular arrangements of cells are regulated by various cell adhesion molecules. On the other hand, cell shape is also known to be regulated by physical constraints similar to those of soap bubbles. In particular, circumference minimization plays an important role, and cell adhesion negatively affects this process, thereby regulating tissue morphogenesis based on physical properties. Here, we focus on the Drosophila compound eye and the mouse auditory epithelium, and summarize the mechanisms of tile pattern formation by cell adhesion molecules such as cadherins, Irre Cell Recognition Modules (IRMs), and nectins. Phenomena that cannot be explained by physical stability based on cortical tension alone have been reported in the tile pattern formation in the compound eye, suggesting that previously unexplored forces such as cellular concentric expansion force may play an important role. We would like to summarize perspectives for future research on the mechanisms of tissue morphogenesis.


Assuntos
Moléculas de Adesão Celular , Sabões , Animais , Camundongos , Adesão Celular/fisiologia , Moléculas de Adesão Celular/metabolismo , Caderinas/metabolismo , Morfogênese/fisiologia , Drosophila/metabolismo
7.
Dev Cell ; 59(1): 156-172.e7, 2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38103554

RESUMO

During morphogenesis, mechanical forces induce large-scale deformations; yet, how forces emerge from cellular contractility and adhesion is unclear. In Drosophila embryos, a tissue-scale wave of actomyosin contractility coupled with adhesion to the surrounding vitelline membrane drives polarized tissue invagination. We show that this process emerges subcellularly from the mechanical coupling between myosin II activation and sequential adhesion/de-adhesion to the vitelline membrane. At the wavefront, integrin clusters anchor the actin cortex to the vitelline membrane and promote activation of myosin II, which in turn enhances adhesion in a positive feedback. Following cell detachment, cortex contraction and advective flow amplify myosin II. Prolonged contact with the vitelline membrane prolongs the integrin-myosin II feedback, increases integrin adhesion, and thus slows down cell detachment and wave propagation. The angle of cell detachment depends on adhesion strength and sets the tensile forces required for detachment. Thus, we document how the interplay between subcellular mechanochemical feedback and geometry drives tissue morphogenesis.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Actomiosina/metabolismo , Miosina Tipo II/metabolismo , Integrinas/metabolismo , Morfogênese/fisiologia
8.
Dev Cell ; 59(2): 262-279.e6, 2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38134928

RESUMO

Organ size is controlled by numerous factors including mechanical forces, which are mediated in part by the Hippo pathway. In growing Drosophila epithelial tissues, cytoskeletal tension influences Hippo signaling by modulating the localization of key pathway proteins to different apical domains. Here, we discovered a Hippo signaling hub at basal spot junctions, which form at the basal-most point of the lateral membranes and resemble adherens junctions in protein composition. Basal spot junctions recruit the central kinase Warts via Ajuba and E-cadherin, which prevent Warts activation by segregating it from upstream Hippo pathway proteins. Basal spot junctions are prominent when tissues undergo morphogenesis and are highly sensitive to fluctuations in cytoskeletal tension. They are distinct from focal adhesions, but the latter profoundly influences basal spot junction abundance by modulating the basal-medial actomyosin network and tension experienced by spot junctions. Thus, basal spot junctions couple morphogenetic forces to Hippo pathway activity and organ growth.


Assuntos
Proteínas de Drosophila , Verrugas , Animais , Drosophila/metabolismo , Via de Sinalização Hippo , Proteínas de Drosophila/metabolismo , Transdução de Sinais , Junções Aderentes/metabolismo , Verrugas/metabolismo , Morfogênese/fisiologia
9.
Methods Mol Biol ; 2745: 177-188, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38060186

RESUMO

Stromal-epithelial interactions mediate mammary gland development and the formation and progression of breast cancer. To study these interactions in vitro, 3D models are essential. We have successfully developed novel 3D in vitro models that allow the formation of mammary gland structures closely resembling those found in vivo and that respond to the hormonal cues that regulate mammary gland morphogenesis and function. Due to their simplicity when compared to in vivo studies, and to their accessibility to visualization in real time, these models are well suited to conceptual and mathematical modeling.


Assuntos
Neoplasias da Mama , Mama , Humanos , Animais , Feminino , Organogênese/fisiologia , Glândulas Mamárias Animais , Morfogênese/fisiologia , Células Epiteliais
10.
C R Biol ; 346: 117-126, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38095130

RESUMO

Morphogenesis, wound healing, and some cancer metastases rely on the collective migration of groups of cells. In these processes, guidance and coordination between cells and tissues are critical. While strongly adherent epithelial cells have to move collectively, loosely organized mesenchymal cells can migrate as individual cells. Nevertheless, many of them migrate collectively. This article summarizes how migratory reactions to cell-cell contacts, also called "contact regulation of locomotion" behaviors, organize mesenchymal collective cell migration. It focuses on one recently discovered mechanism called "guidance by followers", through which a cell is oriented by its immediate followers. In the gastrulating zebrafish embryo, during embryonic axis elongation, this phenomenon is responsible for the collective migration of the leading tissue, the polster, and its guidance by the following posterior axial mesoderm. Such guidance of migrating cells by followers ensures long-range coordination of movements and developmental robustness. Along with other "contact regulation of locomotion" behaviors, this mechanism contributes to organizing collective migration of loose populations of cells.


La morphogénèse, la cicatrisation et certains types de métastases reposent sur la migration collective de groupes de cellules. Lors de ces processus, le guidage et la coordination entre cellules et entre tissus sont fondamentaux. Là où les tissus épithéliaux, très adhésifs, doivent se déplacer collectivement, les cellules mésenchymateuses, en ordre lâche, peuvent migrer individuellement. Cependant, de nombreuses cellules mésenchymateuses migrent de manière collective. Cet article résume comment les réactions migratoires aux contacts entre cellules, aussi appelées «  régulation de locomotion par contact  ¼ , organisent la migration collective des cellules mésenchymateuses. Il décrit en particulier un mécanisme récemment découvert, le «  guidage par les suiveuses  ¼ , par lequel une cellule est orientée par les suiveuses immédiatement en contact. Dans l'embryon de poisson-zèbre en gastrulation, lors de l'élongation du mésoderme axial, ce phénomène est responsable de la migration collective du tissu au front, le polster, et de son guidage par le tissu qui le suit, le mésoderme axial postérieur. Ce mécanisme de guidage par les suiveuses garantit la coordination des mouvements sur de longues distances ainsi que la robustesse du développement. Avec les autres processus de «  régulation de locomotion par contact  ¼ , ce mécanisme contribue à organiser la migration de groupe de cellules en ordre lâche.


Assuntos
Células Epiteliais , Peixe-Zebra , Animais , Movimento Celular/fisiologia , Morfogênese/fisiologia
11.
Curr Biol ; 33(24): 5390-5403.e3, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-37995694

RESUMO

Cell and tissue reshaping is crucial for coordinating three-dimensional pattern formation, in which the size and shape of the cells must be accurately regulated via signal transport and communication among tissues. However, the identity of signaling and transportation mechanisms in this process remains elusive. In our study, we identified an extracellular matrix (ECM) structure with a vertebra-like shape surrounding the central notochord tissue in the larval tail of the urochordate Ciona. Additionally, we verified that the ECM structure was formed de novo, mainly from collagens secreted by notochord cells. Fluorescence recovery after photobleaching and simulation results revealed that this structure was formed via diffusional collagen flow from a notochord that was restricted and molded in the spaces among tail tissues. We revealed that the collagen structure was essential for notochord cell arrangement and elongation. Furthermore, we observed that the central notochord connects with the epidermis through this ECM structure. The disruption of this structure by collagen knockdown and loss-of-collagen function caused the failure of notochord elongation. More importantly, the epidermis could not elongate proportionally with notochord, indicating that the collagen-rich structure serves as a scaffold to coordinate the concurrent elongation of the tail tissues. These findings provide insights into how the central tissue forms and molds its surrounding ECM structure, by not only regulating its own morphogenesis but also functioning as a scaffold for signal transmission to orchestrate the coordinated morphologic reshaping of the surrounding tissues.


Assuntos
Notocorda , Urocordados , Animais , Colágeno , Morfogênese/fisiologia , Matriz Extracelular
12.
Proc Natl Acad Sci U S A ; 120(44): e2305375120, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37871208

RESUMO

Plastic deformation in cells and tissues has been found to play crucial roles in collective cell migration, cancer metastasis, and morphogenesis. However, the fundamental question of how plasticity is initiated in individual cells and then propagates within the tissue remains elusive. Here, we develop a mechanism-based theory of cellular and tissue plasticity that accounts for all key processes involved, including the activation and development of active contraction at different scales as well as the formation of endocytic vesicles on cell junctions and show that this theory achieves quantitative agreement with all existing experiments. Specifically, it reveals that, in response to optical or mechanical stimuli, the myosin contraction and thermal fluctuation-assisted formation and pinching of endocytic vesicles could lead to permanent shortening of cell junctions and that such plastic constriction can stretch neighboring cells and trigger their active contraction through mechanochemical feedbacks and eventually their plastic deformations as well. Our theory predicts that endocytic vesicles with a size around 1 to 2 µm will most likely be formed and a higher irreversible shortening of cell junctions could be achieved if a long stimulation is split into multiple short ones, all in quantitative agreement with experiments. Our analysis also shows that constriction of cells in tissue can undergo elastic/unratcheted to plastic/ratcheted transition as the magnitude and duration of active contraction increases, ultimately resulting in the propagation of plastic deformation waves within the monolayer with a constant speed which again is consistent with experimental observations.


Assuntos
Junções Intercelulares , Morfogênese/fisiologia , Movimento Celular/fisiologia
13.
Elife ; 122023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37782009

RESUMO

In order to understand morphogenesis, it is necessary to know the material properties or forces shaping the living tissue. In spite of this need, very few in vivo measurements are currently available. Here, using the early Drosophila embryo as a model, we describe a novel cantilever-based technique which allows for the simultaneous quantification of applied force and tissue displacement in a living embryo. By analyzing data from a series of experiments in which embryonic epithelium is subjected to developmentally relevant perturbations, we conclude that the response to applied force is adiabatic and is dominated by elastic forces and geometric constraints, or system size effects. Crucially, computational modeling of the experimental data indicated that the apical surface of the epithelium must be softer than the basal surface, a result which we confirmed experimentally. Further, we used the combination of experimental data and comprehensive computational model to estimate the elastic modulus of the apical surface and set a lower bound on the elastic modulus of the basal surface. More generally, our investigations revealed important general features that we believe should be more widely addressed when quantitatively modeling tissue mechanics in any system. Specifically, different compartments of the same cell can have very different mechanical properties; when they do, they can contribute differently to different mechanical stimuli and cannot be merely averaged together. Additionally, tissue geometry can play a substantial role in mechanical response, and cannot be neglected.


Assuntos
Drosophila melanogaster , Drosophila , Animais , Epitélio/fisiologia , Morfogênese/fisiologia , Drosophila melanogaster/metabolismo , Embrião não Mamífero , Modelos Biológicos
14.
Phys Biol ; 20(6)2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37678266

RESUMO

Cells communicate with each other to jointly regulate cellular processes during cellular differentiation and tissue morphogenesis. This multiscale coordination arises through the spatiotemporal activity of morphogens to pattern cell signaling and transcriptional factor activity. This coded information controls cell mechanics, proliferation, and differentiation to shape the growth and morphogenesis of organs. While many of the molecular components and physical interactions have been identified in key model developmental systems, there are still many unresolved questions related to the dynamics involved due to challenges in precisely perturbing and quantitatively measuring signaling dynamics. Recently, a broad range of synthetic optogenetic tools have been developed and employed to quantitatively define relationships between signal transduction and downstream cellular responses. These optogenetic tools can control intracellular activities at the single cell or whole tissue scale to direct subsequent biological processes. In this brief review, we highlight a selected set of studies that develop and implement optogenetic tools to unravel quantitative biophysical mechanisms for tissue growth and morphogenesis across a broad range of biological systems through the manipulation of morphogens, signal transduction cascades, and cell mechanics. More generally, we discuss how optogenetic tools have emerged as a powerful platform for probing and controlling multicellular development.


Assuntos
Fenômenos Biológicos , Optogenética , Morfogênese/fisiologia , Comunicação Celular , Transdução de Sinais/fisiologia
15.
Nature ; 620(7974): 615-624, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37558872

RESUMO

The concomitant occurrence of tissue growth and organization is a hallmark of organismal development1-3. This often means that proliferating and differentiating cells are found at the same time in a continuously changing tissue environment. How cells adapt to architectural changes to prevent spatial interference remains unclear. Here, to understand how cell movements that are key for growth and organization are orchestrated, we study the emergence of photoreceptor neurons that occur during the peak of retinal growth, using zebrafish, human tissue and human organoids. Quantitative imaging reveals that successful retinal morphogenesis depends on the active bidirectional translocation of photoreceptors, leading to a transient transfer of the entire cell population away from the apical proliferative zone. This pattern of migration is driven by cytoskeletal machineries that differ depending on the direction: microtubules are exclusively required for basal translocation, whereas actomyosin is involved in apical movement. Blocking the basal translocation of photoreceptors induces apical congestion, which hampers the apical divisions of progenitor cells and leads to secondary defects in lamination. Thus, photoreceptor migration is crucial to prevent competition for space, and to allow concurrent tissue growth and lamination. This shows that neuronal migration, in addition to its canonical role in cell positioning4, can be involved in coordinating morphogenesis.


Assuntos
Movimento Celular , Morfogênese , Células Fotorreceptoras , Retina , Animais , Humanos , Actomiosina/metabolismo , Competição entre as Células , Diferenciação Celular , Movimento Celular/fisiologia , Proliferação de Células , Microtúbulos/metabolismo , Morfogênese/fisiologia , Organoides/citologia , Organoides/embriologia , Células Fotorreceptoras/citologia , Células Fotorreceptoras/fisiologia , Retina/citologia , Retina/embriologia , Peixe-Zebra/embriologia
16.
J Cell Biol ; 222(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37367826

RESUMO

Branching morphogenesis is an evolutionary solution to maximize epithelial function in a compact organ. It involves successive rounds of branch elongation and branch point formation to generate a tubular network. In all organs, branch points can form by tip splitting, but it is unclear how tip cells coordinate elongation and branching. Here, we addressed these questions in the embryonic mammary gland. Live imaging revealed that tips advance by directional cell migration and elongation relies upon differential cell motility that feeds a retrograde flow of lagging cells into the trailing duct, supported by tip proliferation. Tip bifurcation involved localized repression of cell cycle and cell motility at the branch point. Cells in the nascent daughter tips remained proliferative but changed their direction to elongate new branches. We also report the fundamental importance of epithelial cell contractility for mammary branching morphogenesis. The co-localization of cell motility, non-muscle myosin II, and ERK activities at the tip front suggests coordination/cooperation between these functions.


Assuntos
Células Epiteliais , Glândulas Mamárias Animais , Morfogênese , Divisão Celular , Movimento Celular , Glândulas Mamárias Animais/embriologia , Morfogênese/fisiologia , Mamíferos , Miosina Tipo II/fisiologia
17.
J Cell Biol ; 222(9)2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37351566

RESUMO

Apical constriction is a cell shape change that drives key morphogenetic events during development, including gastrulation and neural tube formation. The forces driving apical constriction are primarily generated through the contraction of apicolateral and/or medioapical actomyosin networks. In the Drosophila ventral furrow, the medioapical actomyosin network has a sarcomere-like architecture, with radially polarized actin filaments and centrally enriched non-muscle myosin II and myosin activating kinase. To determine if this is a broadly conserved actin architecture driving apical constriction, we examined actomyosin architecture during C. elegans gastrulation, in which two endodermal precursor cells internalize from the surface of the embryo. Quantification of protein localization showed that neither the non-muscle myosin II NMY-2 nor the myosin-activating kinase MRCK-1 is enriched at the center of the apex. Further, visualization of barbed- and pointed-end capping proteins revealed that actin filaments do not exhibit radial polarization at the apex. Our results demonstrate that C. elegans endodermal precursor cells apically constrict using a mixed-polarity actin filament network and with myosin and a myosin activator distributed throughout the network. Taken together with observations made in other organisms, our results demonstrate that diverse actomyosin architectures are used in animal cells to accomplish apical constriction.


Assuntos
Actomiosina , Caenorhabditis elegans , Animais , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Caenorhabditis elegans/metabolismo , Constrição , Morfogênese/fisiologia , Miosina Tipo II/metabolismo , Miosinas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo
18.
Nat Commun ; 14(1): 3422, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37296120

RESUMO

The mechanisms that regulate the patterning of branched epithelia remain a subject of long-standing debate. Recently, it has been proposed that the statistical organization of multiple ductal tissues can be explained through a local self-organizing principle based on the branching-annihilating random walk (BARW) in which proliferating tips drive a process of ductal elongation and stochastic bifurcation that terminates when tips encounter maturing ducts. Here, applied to mouse salivary gland, we show the BARW model struggles to explain the large-scale organization of tissue. Instead, we propose that the gland develops as a tip-driven branching-delayed random walk (BDRW). In this framework, a generalization of the BARW, tips inhibited through steric interaction with proximate ducts may continue their branching program as constraints become alleviated through the persistent expansion of the surrounding tissue. This inflationary BDRW model presents a general paradigm for branching morphogenesis when the ductal epithelium grows cooperatively with the domain into which it expands.


Assuntos
Glândulas Salivares , Camundongos , Animais , Epitélio , Morfogênese/fisiologia
19.
Biol Rev Camb Philos Soc ; 98(5): 1749-1767, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37171117

RESUMO

Cranial bones constitute a protective shield for the vulnerable brain tissue, bound together as a rigid entity by unique immovable joints known as sutures. Cranial sutures serve as major growth centres for calvarial morphogenesis and have been identified as a niche for mesenchymal stem cells (MSCs) and/or skeletal stem cells (SSCs) in the craniofacial skeleton. Despite the established dogma of cranial bone and suture biology, technological advancements now allow us to investigate these tissues and structures at unprecedented resolution and embrace multiple novel biological insights. For instance, a decrease or imbalance of representation of SSCs within sutures might underlie craniosynostosis; dural sinuses enable neuroimmune crosstalk and are newly defined as immune hubs; skull bone marrow acts as a myeloid cell reservoir for the meninges and central nervous system (CNS) parenchyma in mediating immune surveillance, etc. In this review, we revisit a growing body of recent studies that explored cranial bone and suture biology using cutting-edge techniques and have expanded our current understanding of this research field, especially from the perspective of development, homeostasis, injury repair, resident MSCs/SSCs, immunosurveillance at the brain's border, and beyond.


Assuntos
Craniossinostoses , Crânio , Humanos , Suturas Cranianas/metabolismo , Craniossinostoses/metabolismo , Morfogênese/fisiologia , Suturas
20.
Dev Growth Differ ; 65(5): 272-281, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37190873

RESUMO

We review several mathematical models and concepts in developmental biology that have been established over the last decade. (1) Feedback vertex set: Ascidian embryos contain cells of seven types, and cell fate is controlled by ~100 interacting genes. The "feedback vertex set" of the directed graph of the gene regulatory network consists of a small number of genes. By experimentally manipulating them, we can differentiate cells into any cell type. (2) Tissue deformation: Describing morphological changes in tissues and relating them to gene expression and other cellular processes is key in understanding morphogenesis. Expansion and anisotropy of the tissue are described by a "deformation tensor" at each location. A study on chick limb bud formation revealed that both the volume growth rate and anisotropy in deformation differed significantly between locations and stages. (3) Mechanobiology: Forces operating on each cell may alter cell shape and gene expression, which may subsequently exert forces on their surroundings. Measurements of force, tissue shape, and gene expression help us understand autonomous tissue deformation. (4) Adaptive design of development: An optimal growth schedule in fluctuating environments explains the growth response to starvation in Drosophila larvae. Adaptive placement of morphogen sources makes development robust to noises.


Assuntos
Drosophila , Organogênese , Animais , Morfogênese/fisiologia , Diferenciação Celular , Organogênese/fisiologia , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...