Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Microsc ; 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37648214

ABSTRACT

Open access to data underpinning published results is a key pillar of scientific reproducibility. Making data available at scale also provides opportunities for data reuse, encouraging the development of new analysis approaches. In this poster article, accompanying a recorded talk, we will explain the benefits of publicly archiving your image data alongside your published manuscripts, as well as highlight what resources are available to do this. This will include the BioImage Archive, EMBL-EBI's new resource for biological image data, https://www.ebi.ac.uk/bioimage-archive/. We will look at how image data submission works, how to prepare in advance for archiving your data and upcoming developments.

2.
iScience ; 25(11): 105330, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36325066

ABSTRACT

Metastasis is tightly linked with poor cancer prognosis, yet it is not clear how transformed cells become invasive carcinomas. We previously discovered that single KRasV12-transformed cells can invade directly from the epithelium by basal cell extrusion. During this process, cells de-differentiate by mechanically pinching off their epithelial determinants, but how they trans-differentiate into a migratory, mesenchymal phenotype is not known. Here, we demonstrate that basally extruded KRasV12-expressing cells become significantly deformed as they invade the zebrafish body. Decreasing the confinement that cells experience after they invade reduces the percentage of KRasV12 cells that trans-differentiate into mesenchymal cell types, while higher confinement increases this percentage. Additionally, increased confinement promotes accumulation of internal masses over time. Altogether, our results suggest that mechanical forces drive not only de-differentiation of KRasV12-transformed epithelial cells as they invade but also their re-differentiation into mesenchymal phenotypes that contribute to distant metastases.

3.
Front Cell Dev Biol ; 10: 947444, 2022.
Article in English | MEDLINE | ID: mdl-36238688

ABSTRACT

Septate junctions (SJs) serve as occluding barriers in invertebrate epithelia. In Drosophila, at least 30 genes are required for the formation or maintenance of SJs. Interestingly, loss-of-function mutations in core SJ components are embryonic lethal, with defects in developmental events such as head involution and dorsal closure (DC) that occur prior to the formation of a mature SJ, indicating a role for these proteins in mid-embryogenesis independent of their occluding function. To understand this novel function in development, we examined loss-of-function mutations in three core SJ proteins during the process of DC. DC occurs during mid-embryogenesis to seal a dorsal gap in the epidermis following germ band retraction. Closure is driven by contraction of the extraembryonic amnioserosa cells that temporarily cover the dorsal surface and by cell shape changes (elongation) of lateral epidermal cells that bring the contralateral sheets together at the dorsal midline. Using live imaging and examination of fixed tissues, we show that early events in DC occur normally in SJ mutant embryos, but during later closure, coracle, Macroglobulin complement-related and Neurexin-IV mutant embryos exhibit slower rates of closure and display aberrant cells shapes in the dorsolateral epidermis, including dorsoventral length and apical surface area. SJ mutant embryos also show mild defects in actomyosin structures along the leading edge, but laser cutting experiments suggest similar tension and viscoelastic properties in SJ mutant versus wild type epidermis. In a high percentage of SJ mutant embryos, the epidermis tears free from the amnioserosa near the end of DC and live imaging and immunostaining reveal reduced levels of E-cadherin, suggesting that defective adhesion may be responsible for these tears. Supporting this notion, reducing E-cadherin by half significantly enhances the penetrance of DC defects in coracle mutant embryos.

4.
Curr Opin Genet Dev ; 72: 1-7, 2022 02.
Article in English | MEDLINE | ID: mdl-34560388

ABSTRACT

Epithelia remove dying or excess cells by extrusion, a process that seamlessly squeezes cells out of the layer without disrupting their barrier function. New studies shed light into the intricate relationship between extrusion, tissue mechanics, and development. They emphasize the importance of whole tissue-mechanics, rather than single cell-mechanics in controlling extrusion. Tissue compaction, stiffness, and cell-cell adhesion can impact the efficiency of cell extrusion and mechanisms that drive it, to adapt to different conditions during development or disease.


Subject(s)
Apoptosis , Epithelial Cells , Epithelium
5.
Nat Commun ; 12(1): 7180, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893591

ABSTRACT

Metastasis is the main cause of carcinoma-related death, yet we know little about how it initiates due to our inability to visualize stochastic invasion events. Classical models suggest that cells accumulate mutations that first drive formation of a primary mass, and then downregulate epithelia-specific genes to cause invasion and metastasis. Here, using transparent zebrafish epidermis to model simple epithelia, we can directly image invasion. We find that KRas-transformation, implicated in early carcinogenesis steps, directly drives cell invasion by hijacking a process epithelia normally use to promote death-cell extrusion. Cells invading by basal cell extrusion simultaneously pinch off their apical epithelial determinants, endowing new plasticity. Following invasion, cells divide, enter the bloodstream, and differentiate into stromal, neuronal-like, and other cell types. Yet, only invading KRasV12 cells deficient in p53 survive and form internal masses. Together, we demonstrate that KRas-transformation alone causes cell invasion and partial dedifferentiation, independently of mass formation.


Subject(s)
Epithelial Cells/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Movement , Epidermis/metabolism , Epithelium/metabolism , Humans , Neoplasms/diagnostic imaging , Zebrafish/metabolism , Zebrafish Proteins
6.
Development ; 144(23): 4298-4312, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28870989

ABSTRACT

How position-dependent cell fate acquisition occurs during embryogenesis is a central question in developmental biology. To study this process, we developed a defined, high-throughput assay to induce peri-gastrulation-associated patterning in geometrically confined human pluripotent stem cell (hPSC) colonies. We observed that, upon BMP4 treatment, phosphorylated SMAD1 (pSMAD1) activity in the colonies organized into a radial gradient. We developed a reaction-diffusion (RD)-based computational model and observed that the self-organization of pSMAD1 signaling was consistent with the RD principle. Consequent fate acquisition occurred as a function of both pSMAD1 signaling strength and duration of induction, consistent with the positional-information (PI) paradigm. We propose that the self-organized peri-gastrulation-like fate patterning in BMP4-treated geometrically confined hPSC colonies arises via a stepwise model of RD followed by PI. This two-step model predicted experimental responses to perturbations of key parameters such as colony size and BMP4 dose. Furthermore, it also predicted experimental conditions that resulted in RD-like periodic patterning in large hPSC colonies, and rescued peri-gastrulation-like patterning in colony sizes previously thought to be reticent to this behavior.


Subject(s)
Body Patterning/physiology , Gastrulation/physiology , Models, Biological , Body Patterning/genetics , Bone Morphogenetic Protein 4/physiology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Differentiation/physiology , Cells, Cultured , Colony-Forming Units Assay/methods , Gastrulation/genetics , High-Throughput Screening Assays/methods , Humans , Nodal Protein/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , RNA, Small Interfering/genetics , Signal Transduction , Smad1 Protein/physiology
7.
Curr Biol ; 27(15): 2260-2270.e5, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28736167

ABSTRACT

In response to a pulling force, a material can elongate, hold fast, or fracture. During animal development, multi-cellular contraction of one region often stretches neighboring tissue. Such local contraction occurs by induced actomyosin activity, but molecular mechanisms are unknown for regulating the physical properties of connected tissue for elongation under stress. We show that cytohesins, and their Arf small G protein guanine nucleotide exchange activity, are required for tissues to elongate under stress during both Drosophila dorsal closure (DC) and zebrafish epiboly. In Drosophila, protein localization, laser ablation, and genetic interaction studies indicate that the cytohesin Steppke reduces tissue tension by inhibiting actomyosin activity at adherens junctions. Without Steppke, embryogenesis fails, with epidermal distortions and tears resulting from myosin misregulation. Remarkably, actomyosin network assembly is necessary and sufficient for local Steppke accumulation, where live imaging shows Steppke recruitment within minutes. This rapid negative feedback loop provides a molecular mechanism for attenuating the main tension generator of animal tissues. Such attenuation relaxes tissues and allows orderly elongation under stress.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , GTP-Binding Protein Regulators/genetics , Guanine Nucleotide Exchange Factors/genetics , Signal Transduction , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , GTP-Binding Protein Regulators/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
8.
Mol Biol Cell ; 28(10): 1301-1310, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28331071

ABSTRACT

Tissue morphogenesis relies on the coordinated action of actin networks, cell-cell adhesions, and cell-extracellular matrix (ECM) adhesions. Such coordination can be achieved through cross-talk between cell-cell and cell-ECM adhesions. Drosophila dorsal closure (DC), a morphogenetic process in which an extraembryonic tissue called the amnioserosa contracts and ingresses to close a discontinuity in the dorsal epidermis of the embryo, requires both cell-cell and cell-ECM adhesions. However, whether the functions of these two types of adhesions are coordinated during DC is not known. Here we analyzed possible interdependence between cell-cell and cell-ECM adhesions during DC and its effect on the actomyosin network. We find that loss of cell-ECM adhesion results in aberrant distributions of cadherin-mediated adhesions and actin networks in the amnioserosa and subsequent disruption of myosin recruitment and dynamics. Moreover, loss of cell-cell adhesion caused up-regulation of cell-ECM adhesion, leading to reduced cell deformation and force transmission across amnioserosa cells. Our results show how interdependence between cell-cell and cell-ECM adhesions is important in regulating cell behaviors, force generation, and force transmission critical for tissue morphogenesis.


Subject(s)
Cell Adhesion/physiology , Cell-Matrix Junctions/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Actomyosin/metabolism , Animals , Cadherins/metabolism , Cell Movement/physiology , Drosophila/embryology , Drosophila/metabolism , Drosophila Proteins/metabolism , Embryonic Development/physiology , Epidermis/metabolism , Extracellular Matrix/metabolism , Myosins/metabolism
9.
J Cell Sci ; 130(4): 689-696, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28202603

ABSTRACT

Embryos repair epithelial wounds rapidly in a process driven by collective cell movements. Upon wounding, actin and the molecular motor non-muscle myosin II are redistributed in the cells adjacent to the wound, forming a supracellular purse string around the lesion. Purse string contraction coordinates cell movements and drives rapid wound closure. By using fluorescence recovery after photobleaching in Drosophila embryos, we found that myosin turns over as the purse string contracts. Myosin turnover at the purse string was slower than in other actomyosin networks that had a lower level of contractility. Mathematical modelling suggested that myosin assembly and disassembly rates were both reduced by tension at the wound edge. We used laser ablation to show that tension at the purse string increased as wound closure progressed, and that the increase in tension was associated with reduced myosin turnover. Reducing purse string tension by laser-mediated severing resulted in increased turnover and loss of myosin. Finally, myosin motor activity was necessary for its stabilization around the wound and for rapid wound closure. Our results indicate that mechanical forces regulate myosin dynamics during embryonic wound repair.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Myosin Type II/metabolism , Wound Healing , Animals , Biomechanical Phenomena , Phosphorylation , Protein Stability
10.
Mech Dev ; 144(Pt A): 43-52, 2017 04.
Article in English | MEDLINE | ID: mdl-27989746

ABSTRACT

Embryonic tissues display an outstanding ability to rapidly repair wounds. Epithelia, in particular, serve as protective layers that line internal organs and form the skin. Thus, maintenance of epithelial integrity is of utmost importance for animal survival, particularly at embryonic stages, when an immune system has not yet fully developed. Rapid embryonic repair of epithelial tissues is conserved across species, and involves the collective migration of the cells around the wound. The migratory cell behaviours associated with wound repair require the generation and transmission of mechanical forces, not only for the cells to move, but also to coordinate their movements. Here, we review the forces involved in embryonic wound repair. We discuss how different force-generating structures are assembled at the molecular level, and the mechanisms that maintain the balance between force-generating structures as wounds close. Finally, we describe the mechanisms that cells use to coordinate the generation of mechanical forces around the wound. Collective cell movements and their misregulation have been associated with defective tissue repair, developmental abnormalities and cancer metastasis. Thus, we propose that understanding the role of mechanical forces during embryonic wound closure will be crucial to develop therapeutic interventions that promote or prevent collective cell movements under pathological conditions.


Subject(s)
Drosophila melanogaster/embryology , Embryonic Development/genetics , Epidermis/embryology , Gene Expression Regulation, Developmental , Wound Healing/genetics , Actins/genetics , Actins/metabolism , Adherens Junctions/metabolism , Animals , Biomechanical Phenomena , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Epidermal Cells , Epidermis/metabolism , Models, Biological , Myosins/genetics , Myosins/metabolism , Signal Transduction
11.
Dev Cell ; 39(5): 611-625, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27923121

ABSTRACT

Tissue morphogenesis requires force-generating mechanisms to organize cells into complex structures. Although many such mechanisms have been characterized, we know little about how forces are integrated across developing tissues. We provide evidence that integrin-mediated cell-extracellular matrix (ECM) adhesion modulates the transmission of apically generated tension during dorsal closure (DC) in Drosophila. Integrin-containing adhesive structures resembling focal adhesions were identified on the basal surface of the amnioserosa (AS), an extraembryonic epithelium essential for DC. Genetic modulation of integrin-mediated adhesion results in defective DC. Quantitative image analysis and laser ablation experiments reveal that basal cell-ECM adhesions provide resistance to apical cell displacements and force transmission between neighboring cells in the AS. Finally, we provide evidence for integrin-dependent force transmission to the AS substrate. Overall, we find that integrins regulate force transmission within and between cells, thereby playing an essential role in transmitting tension in developing tissues.


Subject(s)
Drosophila/embryology , Animals , Animals, Genetically Modified , Biophysical Phenomena , Cell Adhesion/physiology , Drosophila/cytology , Drosophila/physiology , Drosophila Proteins/physiology , Extracellular Matrix/physiology , Focal Adhesions/physiology , Integrins/physiology , Models, Biological , Morphogenesis/physiology
12.
Integr Biol (Camb) ; 7(3): 298-312, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25609037

ABSTRACT

During development and in adult tissues separation of phenotypically distinct cell populations is necessary to ensure proper organization and function of tissues and organs. Various phenomena, such as differential adhesion, differential mechanical tension and cell-cell repulsion, are proposed to cause boundary formation. Moreover, emerging evidence suggests that interplay between multiple such phenomena can underlie boundary formation. Boundary-forming mechanisms are commonly studied in vivo in complex embryo models or in vitro using simple model systems not reflective of in vivo boundary complexity. To better elucidate the interplay between multiple boundary formation mechanism, there is therefore a need for more relevant in vitro model systems that allow quantitative and concomitant studies of the multiple changes in cell/tissue behaviour that lead to boundary establishment. Here, we develop such a model using patterned co-cultures of two cell populations. Using a set of quantitative tools, we demonstrate that our approach allows us to study the mechanisms underlying boundary formation. We demonstrate that in our specific system differential mechanical tension and modulation of migratory behavior of cells accompany boundary formation. The design of our in vitro model system will allow researchers to obtain quantitative, integrative mechanistic data facilitating a faster and more thorough understanding of the fundamental principles underlying boundary formation.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Focal Adhesions/physiology , Membrane Fluidity/physiology , Morphogenesis/physiology , Cell Adhesion/physiology , Cell Enlargement , Cell Line , Cell Movement/physiology , Cell Size , Coculture Techniques/methods , Humans , Models, Biological , Stress, Mechanical
13.
Development ; 141(14): 2901-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24948602

ABSTRACT

The embryonic epidermis displays a remarkable ability to repair wounds rapidly. Embryonic wound repair is driven by the evolutionary conserved redistribution of cytoskeletal and junctional proteins around the wound. Drosophila has emerged as a model to screen for factors implicated in wound closure. However, genetic screens have been limited by the use of manual analysis methods. We introduce MEDUSA, a novel image-analysis tool for the automated quantification of multicellular and molecular dynamics from time-lapse confocal microscopy data. We validate MEDUSA by quantifying wound closure in Drosophila embryos, and we show that the results of our automated analysis are comparable to analysis by manual delineation and tracking of the wounds, while significantly reducing the processing time. We demonstrate that MEDUSA can also be applied to the investigation of cellular behaviors in three and four dimensions. Using MEDUSA, we find that the conserved nonreceptor tyrosine kinase Abelson (Abl) contributes to rapid embryonic wound closure. We demonstrate that Abl plays a role in the organization of filamentous actin and the redistribution of the junctional protein ß-catenin at the wound margin during embryonic wound repair. Finally, we discuss different models for the role of Abl in the regulation of actin architecture and adhesion dynamics at the wound margin.


Subject(s)
Automation , Drosophila melanogaster/embryology , Image Processing, Computer-Assisted , Proto-Oncogene Proteins c-abl/metabolism , Wound Healing , Actins/metabolism , Algorithms , Animals , Cell Tracking , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/enzymology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/enzymology , Microscopy, Confocal , Models, Biological , Pseudopodia/metabolism , Reproducibility of Results , beta Catenin/metabolism
14.
Article in English | MEDLINE | ID: mdl-24110601

ABSTRACT

In this article we investigate the suitability of a manifold learning technique to classify different types of emphysema based on embedded Probabilistic PCA (PPCA). Our approach finds the most discriminant linear space for each emphysema pattern against the remaining patterns where lung CT image patches can be embedded. In this embedded space, we train a PPCA model for each pattern. The main novelty of our technique is that it is possible to compute the class membership posterior probability for each emphysema pattern rather than a hard assignment as it is typically done by other approaches. We tested our algorithm with six emphysema patterns using a data set of 1337 CT training patches. Using a 10-fold cross validation experiment, an average recall rate of 69% is achieved when the posterior probability is greater than 75%. A quantitative comparison with a texture-based approach based on Local Binary Patterns and with an approach based on local intensity distributions shows that our method is competitive. The analysis of full lungs using our approach shows a good visual agreement with the underlying emphysema types and a smooth spatial relation.


Subject(s)
Pulmonary Emphysema/diagnostic imaging , Algorithms , Discriminant Analysis , Humans , Lung/diagnostic imaging , Lung/pathology , Principal Component Analysis , Pulmonary Emphysema/classification , Radiographic Image Interpretation, Computer-Assisted , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...