Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
2.
PLoS Biol ; 21(8): e3002261, 2023 08.
Article in English | MEDLINE | ID: mdl-37590318

ABSTRACT

Epithelial-mesenchymal transition (EMT) is an early event in cell dissemination from epithelial tissues. EMT endows cells with migratory, and sometimes invasive, capabilities and is thus a key process in embryo morphogenesis and cancer progression. So far, matrix metalloproteinases (MMPs) have not been considered as key players in EMT but rather studied for their role in matrix remodelling in later events such as cell migration per se. Here, we used Xenopus neural crest cells to assess the role of MMP28 in EMT and migration in vivo. We show that a catalytically active MMP28, expressed by neighbouring placodal cells, is required for neural crest EMT and cell migration. We provide strong evidence indicating that MMP28 is imported in the nucleus of neural crest cells where it is required for normal Twist expression. Our data demonstrate that MMP28 can act as an upstream regulator of EMT in vivo raising the possibility that other MMPs might have similar early roles in various EMT-related contexts such as cancer, fibrosis, and wound healing.


Subject(s)
Epithelial-Mesenchymal Transition , Neural Crest , Cell Movement , Cell Nucleus , Epithelium
4.
Cells Dev ; 168: 203723, 2021 12.
Article in English | MEDLINE | ID: mdl-34284169

ABSTRACT

The neural tube is the precursor of the central nervous system. Its early formation and growth are known to be extremely biased along the anteroposterior (AP) axis. Several mechanisms including addition of cells from the tail bud, lateral pressure from surrounding tissues and oriented cell divisions have been proposed to contribute to this biased growth. Here we show that, contrary to what has been found in posterior regions encompassing the tail bud region, the growth of the anterior trunk neural tube is slower along the AP direction than in the other axes. We found that this is due to anchorage of the neural tube to the matrix which favors apicobasal elongation at the expense of AP growth. In addition, as the neural tube develops, we found a moderate slowdown of cell proliferation that could account for the overall reduction of the pace of 3D growth in the same time window. However, as we found no preferred orientation of cell division, changes in cell cycle pace are unlikely to directly contribute to the observed AP-hindered growth of neural tube. Overall, these data indicate that neural tube growth is not intrinsically positively biased along the AP axis. Rather it switches from AP-favored to AP-hindered regimes between the most posterior and anterior trunk neural tube regions.


Subject(s)
Chickens , Neural Tube , Animals , Cell Division , Central Nervous System , Mesoderm , Neural Tube/metabolism
5.
Methods Mol Biol ; 2179: 257-274, 2021.
Article in English | MEDLINE | ID: mdl-32939726

ABSTRACT

The epithelial-mesenchymal transition (EMT) converts coherent epithelial structures into single cells. EMT is a dynamic cellular process that is not systematically completed (not all EMTs lead to single cells) and reversible (cells can re-epithelialize). EMT is orchestrated at multiple levels from transcription, to posttranslational modifications, to protein turnover. It involves remodeling of polarity and adhesion and enhances migratory capabilities. During physiological events such as embryogenesis or wound healing EMT is used to initiate cell migration, but EMT can also occur in pathological settings. In particular, EMT has been linked to fibrosis and cancer. Neural crest (NC) cells, an embryonic stem cell population whose behavior recapitulates the main steps of carcinoma progression, are a great model to study EMT. In this chapter, we provide a fully detailed protocol to extract NC cells from Xenopus embryos and culture them to study the dynamics of cell-cell adhesion, cell motility, and dispersion.


Subject(s)
Cell Tracking/methods , Epithelial-Mesenchymal Transition , Neural Crest/cytology , Primary Cell Culture/methods , Animals , Cell Adhesion , Cell Movement , Cell Tracking/instrumentation , Xenopus
6.
Front Physiol ; 11: 586432, 2020.
Article in English | MEDLINE | ID: mdl-33324240

ABSTRACT

Directed cell migration is essential all along an individual's life, from embryogenesis to tissue repair and cancer metastasis. Thus, due to its biomedical relevance, directed cell migration is currently under intense research. Directed cell migration has been shown to be driven by an assortment of external biasing cues, ranging from gradients of soluble (chemotaxis) to bound (haptotaxis) molecules. In addition to molecular gradients, gradients of mechanical properties (duro/mechanotaxis), electric fields (electro/galvanotaxis) as well as iterative biases in the environment topology (ratchetaxis) have been shown to be able to direct cell migration. Since cells migrating in vivo are exposed to a challenging environment composed of a convolution of biochemical, biophysical, and topological cues, it is highly unlikely that cell migration would be guided by an individual type of "taxis." This is especially true since numerous molecular players involved in the cellular response to these biasing cues are often recycled, serving as sensor or transducer of both biochemical and biophysical signals. In this review, we confront literature on Xenopus cephalic neural crest cells with that of other cell types to discuss the relevance of the current categorization of cell guidance strategies. Furthermore, we emphasize that while studying individual biasing signals is informative, the hard truth is that cells migrate by performing a sort of "mixotaxis," where they integrate and coordinate multiple inputs through shared molecular effectors to ensure robustness of directed cell motion.

7.
Philos Trans R Soc Lond B Biol Sci ; 375(1809): 20190559, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32829678

ABSTRACT

Matrix metalloproteinases (MMPs) are a large family of proteases comprising 24 members in vertebrates. They are well known for their extracellular matrix remodelling activity. MMP28 is the latest member of the family to be discovered. It is a secreted MMP involved in wound healing, immune system maturation, cell survival and migration. MMP28 is also expressed during embryogenesis in human and mouse. Here, we describe the detailed expression profile of MMP28 in Xenopus laevis embryos. We show that MMP28 is expressed maternally and accumulates at neurula and tail bud stages specifically in the cranial placode territories adjacent to migrating neural crest cells. As a secreted MMP, MMP28 may be required in neural crest-placode interactions. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.


Subject(s)
Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental , Matrix Metalloproteinases/genetics , Morphogenesis , Skull/embryology , Xenopus Proteins/genetics , Xenopus laevis/embryology , Animals , Matrix Metalloproteinases/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism
8.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Article in English | MEDLINE | ID: mdl-32300252

ABSTRACT

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Subject(s)
Biomedical Research/standards , Epithelial-Mesenchymal Transition , Animals , Cell Movement , Cell Plasticity , Consensus , Developmental Biology/standards , Humans , Neoplasms/pathology , Terminology as Topic
9.
Development ; 147(7)2020 04 12.
Article in English | MEDLINE | ID: mdl-32280063

ABSTRACT

Matrix metalloproteinases have a broad spectrum of substrates ranging from extracellular matrix components and adhesion molecules to chemokines and growth factors. Despite being mostly secreted, MMPs have been detected in the cytosol, the mitochondria or the nucleus. Although most of the attention is focused on their role in matrix remodeling, the diversity of their substrates and their complex trafficking open the possibility for non-canonical functions. Yet in vivo examples and experimental demonstration of the physiological relevance of such activities are rare. Here, we have used chick neural crest (NC) cells, a highly migratory stem cell population likened to invasive cancer cells, as a model for physiological epithelial-mesenchymal transition (EMT). We demonstrate that MMP14 is required for NC delamination. Interestingly, this role is independent of its cytoplasmic tail and of its catalytic activity. Our in vivo data indicate that, in addition to being a late pro-invasive factor, MMP14 is also likely to be an early player, owing to its role in EMT.


Subject(s)
Extracellular Matrix/metabolism , Lamin Type A/metabolism , Matrix Metalloproteinase 14/physiology , Neural Crest/metabolism , Animals , Animals, Genetically Modified , Cadherins/metabolism , Catalysis , Cells, Cultured , Chick Embryo , Epithelial-Mesenchymal Transition/physiology
10.
PLoS Comput Biol ; 15(12): e1007171, 2019 12.
Article in English | MEDLINE | ID: mdl-31869321

ABSTRACT

Pseudostratified epithelia (PSE) are a common type of columnar epithelia found in a wealth of embryonic and adult tissues such as ectodermal placodes, the trachea, the ureter, the gut and the neuroepithelium. PSE are characterized by the choreographed displacement of cells' nuclei along the apicobasal axis according to phases of their cell cycle. Such movements, called interkinetic movements (INM), have been proposed to influence tissue expansion and shape and suggested as culprit in several congenital diseases such as CAKUT (Congenital anomalies of kidney and urinary tract) and esophageal atresia. INM rely on cytoskeleton dynamics just as adhesion, contractility and mitosis do. Therefore, long term impairment of INM without affecting proliferation and adhesion is currently technically unachievable. Here we bypassed this hurdle by generating a 2D agent-based model of a proliferating PSE and compared its output to the growth of the chick neuroepithelium to assess the interplay between INM and these other important cell processes during growth of a PSE. We found that INM directly generates apical expansion and apical nuclear crowding. In addition, our data strongly suggest that apicobasal elongation of cells is not an emerging property of a proliferative PSE but rather requires a specific elongation program. We then discuss how such program might functionally link INM, tissue growth and differentiation.


Subject(s)
Cell Nucleus/physiology , Epithelium/embryology , Animals , Body Patterning/physiology , Cell Count , Cell Cycle/physiology , Cell Polarity/physiology , Cell Proliferation/physiology , Chick Embryo , Computational Biology , Humans , Models, Biological , Movement/physiology , Neuroepithelial Cells/cytology , Systems Analysis , Urogenital Abnormalities/embryology , Vesico-Ureteral Reflux/embryology
11.
Nat Commun ; 10(1): 1518, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944331

ABSTRACT

When migrating in vivo, cells are exposed to numerous conflicting signals: chemokines, repellents, extracellular matrix, growth factors. The roles of several of these molecules have been studied individually in vitro or in vivo, but we have yet to understand how cells integrate them. To start addressing this question, we used the cephalic neural crest as a model system and looked at the roles of its best examples of positive and negative signals: stromal-cell derived factor 1 (Sdf1/Cxcl12) and class3-Semaphorins. Here we show that Sdf1 and Sema3A antagonistically control cell-matrix adhesion via opposite effects on Rac1 activity at the single cell level. Directional migration at the population level emerges as a result of global Semaphorin-dependent confinement and broad activation of adhesion by Sdf1 in the context of a biased Fibronectin distribution. These results indicate that uneven in vivo topology renders the need for precise distribution of secreted signals mostly dispensable.


Subject(s)
Cell Movement/physiology , Cell-Matrix Junctions/physiology , Neural Crest/cytology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cell Line , Cell Shape/drug effects , Cell Surface Extensions/drug effects , Cell-Matrix Junctions/drug effects , Cell-Matrix Junctions/metabolism , Chemokine CXCL12/metabolism , Female , Fibronectins/metabolism , Male , Manganese/metabolism , Mice , Nerve Tissue Proteins/physiology , Neural Crest/drug effects , Neural Crest/metabolism , Receptors, CXCR4/metabolism , Semaphorins/metabolism , Xenopus laevis/embryology , rac1 GTP-Binding Protein/metabolism
12.
PLoS Comput Biol ; 15(4): e1007002, 2019 04.
Article in English | MEDLINE | ID: mdl-31009457

ABSTRACT

A fundamental question in embryo morphogenesis is how a complex pattern is established in seemingly uniform tissues. During vertebrate development, neural crest cells differentiate as a continuous mass of tissue along the neural tube and subsequently split into spatially distinct migratory streams to invade the rest of the embryo. How these streams are established is not well understood. Inhibitory signals surrounding the migratory streams led to the idea that position and size of streams are determined by a pre-pattern of such signals. While clear evidence for a pre-pattern in the cranial region is still lacking, all computational models of neural crest migration published so far have assumed a pre-pattern of negative signals that channel the neural crest into streams. Here we test the hypothesis that instead of following a pre-existing pattern, the cranial neural crest creates their own migratory pathway by interacting with the surrounding tissue. By combining theoretical modeling with experimentation, we show that streams emerge from the interaction of the hindbrain neural crest and the neighboring epibranchial placodal tissues, without the need for a pre-existing guidance cue. Our model suggests that the initial collective neural crest invasion is based on short-range repulsion and asymmetric attraction between neighboring tissues. The model provides a coherent explanation for the formation of cranial neural crest streams in concert with previously reported findings and our new in vivo observations. Our results point to a general mechanism of inducing collective invasion patterns.


Subject(s)
Morphogenesis/physiology , Neural Crest/embryology , Animals , Anura/embryology , Cell Adhesion/physiology , Computational Biology , Embryo, Mammalian/embryology , Embryo, Nonmammalian/embryology , Mice , Models, Biological , Rhombencephalon/embryology
13.
Genesis ; 56(6-7): e23107, 2018 06.
Article in English | MEDLINE | ID: mdl-29675839

ABSTRACT

Neural crest (NC) cells were described for the first time in 1868 by Wilhelm His. Since then, this amazing population of migratory stem cells has been intensively studied. It took a century to fully unravel their incredible abilities to contribute to nearly every organ of the body. Yet, our understanding of the cell and molecular mechanisms controlling their migration is far from complete. In this review, we summarize the current knowledge on epithelial-mesenchymal transition and collective behavior of NC cells and propose further stops at which the NC train might be calling in the near future.


Subject(s)
Cell Movement/physiology , Neural Crest/embryology , Animals , Cell Adhesion/physiology , Epithelial-Mesenchymal Transition/physiology , Humans
14.
F1000Res ; 6: 1899, 2017.
Article in English | MEDLINE | ID: mdl-29152225

ABSTRACT

Collective cell migration is the coordinated movement emerging from the interaction of at least two cells. In multicellular organisms, collective cell migration is ubiquitous. During development, embryonic cells often travel in numbers, whereas in adults, epithelial cells close wounds collectively. There is often a division of labour and two categories of cells have been proposed: leaders and followers. These two terms imply that followers are subordinated to leaders whose proposed broad range of actions significantly biases the direction of the group of cells towards a specific target. These two terms are also tied to topology. Leaders are at the front while followers are located behind them. Here, we review recent work on some of the main experimental models for collective cell migration, concluding that leader-follower terminology may not be the most appropriate. It appears that not all collectively migrating groups are driven by cells located at the front. Moreover, the qualities that define leaders (pathfinding, traction forces and matrix remodelling) are not specific to front cells. These observations indicate that the terms leaders and followers are not suited to every case. We think that it would be more accurate to dissociate the function of a cell from its position in the group. The position of cells can be precisely defined with respect to the direction of movement by purely topological terms such as "front" or "rear" cells. In addition, we propose the more ample and strictly functional definition of "steering cells" which are able to determine the directionality of movement for the entire group. In this context, a leader cell represents only a specific case in which a steering cell is positioned at the front of the group.

15.
Development ; 144(13): 2456-2468, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28526750

ABSTRACT

A fundamental property of neural crest (NC) migration is contact inhibition of locomotion (CIL), a process by which cells change their direction of migration upon cell contact. CIL has been proven to be essential for NC migration in amphibians and zebrafish by controlling cell polarity in a cell contact-dependent manner. Cell contact during CIL requires the participation of the cell adhesion molecule N-cadherin, which starts to be expressed by NC cells as a consequence of the switch between E- and N-cadherins during epithelial-to-mesenchymal transition (EMT). However, the mechanism that controls the upregulation of N-cadherin remains unknown. Here, we show that platelet-derived growth factor receptor alpha (PDGFRα) and its ligand platelet-derived growth factor A (PDGF-A) are co-expressed in migrating cranial NC. Inhibition of PDGF-A/PDGFRα blocks NC migration by inhibiting N-cadherin and, consequently, impairing CIL. Moreover, we identify phosphatidylinositol-3-kinase (PI3K)/AKT as a downstream effector of the PDGFRα cellular response during CIL. Our results lead us to propose PDGF-A/PDGFRα signalling as a tissue-autonomous regulator of CIL by controlling N-cadherin upregulation during EMT. Finally, we show that once NC cells have undergone EMT, the same PDGF-A/PDGFRα works as an NC chemoattractant, guiding their directional migration.


Subject(s)
Cadherins/metabolism , Cell Movement , Contact Inhibition , Locomotion , Neural Crest/cytology , Platelet-Derived Growth Factor/metabolism , Xenopus laevis/metabolism , Animals , Cell Movement/drug effects , Chemotaxis/drug effects , Contact Inhibition/drug effects , Locomotion/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology
17.
Elife ; 42015 Oct 13.
Article in English | MEDLINE | ID: mdl-26459831

ABSTRACT

Dystrophin forms an essential link between sarcolemma and cytoskeleton, perturbation of which causes muscular dystrophy. We analysed Dystrophin binding dynamics in vivo for the first time. Within maturing fibres of host zebrafish embryos, our analysis reveals a pool of diffusible Dystrophin and complexes bound at the fibre membrane. Combining modelling, an improved FRAP methodology and direct semi-quantitative analysis of bleaching suggests the existence of two membrane-bound Dystrophin populations with widely differing bound lifetimes: a stable, tightly bound pool, and a dynamic bound pool with high turnover rate that exchanges with the cytoplasmic pool. The three populations were found consistently in human and zebrafish Dystrophins overexpressed in wild-type or dmd(ta222a/ta222a) zebrafish embryos, which lack Dystrophin, and in Gt(dmd-Citrine)(ct90a) that express endogenously-driven tagged zebrafish Dystrophin. These results lead to a new model for Dystrophin membrane association in developing muscle, and highlight our methodology as a valuable strategy for in vivo analysis of complex protein dynamics.


Subject(s)
Dystrophin/analysis , Muscle, Skeletal/chemistry , Muscle, Skeletal/growth & development , Zebrafish/embryology , Animals , Fluorescence Recovery After Photobleaching , Humans
18.
Dev Cell ; 34(4): 421-34, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26235046

ABSTRACT

Contact inhibition of locomotion (CIL) is the process through which cells move away from each other after cell-cell contact, and it contributes to malignant invasion and developmental migration. Various cell types exhibit CIL, whereas others remain in contact after collision and may form stable junctions. To investigate what determines this differential behavior, we study neural crest cells, a migratory stem cell population whose invasiveness has been likened to cancer metastasis. By comparing pre-migratory and migratory neural crest cells, we show that the switch from E- to N-cadherin during EMT is essential for acquisition of CIL behavior. Loss of E-cadherin leads to repolarization of protrusions, via p120 and Rac1, resulting in a redistribution of forces from intercellular tension to cell-matrix adhesions, which break down the cadherin junction. These data provide insight into the balance of physical forces that contributes to CIL in cells in vivo.


Subject(s)
Cadherins/metabolism , Cell Movement , Cell Polarity , Contact Inhibition , Epithelial-Mesenchymal Transition , Neural Crest/cytology , Animals , Biomechanical Phenomena , Catenins/metabolism , Intercellular Junctions/metabolism , Protein Binding , Xenopus laevis , rac1 GTP-Binding Protein/metabolism , Delta Catenin
20.
Med Sci (Paris) ; 30(8-9): 751-7, 2014.
Article in French | MEDLINE | ID: mdl-25174751

ABSTRACT

Historically centered on the study of individual cell motility, the field of cell migration has recently moved up one level to look at cooperative behaviour within migratory cell populations. It is now well established that numerous physiological and pathological migration events involve collectively migrating cells rather than solitary cells or concomitantly migrating individual cells. In this review, we first discuss the criteria allowing a given migratory event to be classified as collective cell migration. We then summarize the main concepts that rule collective cell migration in epithelial and mesenchymal tissues with a main focus on mechanisms controlling polarity and directionality in cell collectives.


Subject(s)
Cell Movement/physiology , Animals , Cell Communication/physiology , Cell Polarity/physiology , Embryonic Development/physiology , Humans , Immunity/physiology , Signal Transduction , Wound Healing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...