Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32300252

RESUMEN

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.


Asunto(s)
Investigación Biomédica/normas , Transición Epitelial-Mesenquimal , Animales , Movimiento Celular , Plasticidad de la Célula , Consenso , Biología Evolutiva/normas , Humanos , Neoplasias/patología , Terminología como Asunto
3.
PLoS Biol ; 21(8): e3002261, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37590318

RESUMEN

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.


Asunto(s)
Transición Epitelial-Mesenquimal , Cresta Neural , Movimiento Celular , Núcleo Celular , Epitelio
4.
Development ; 147(7)2020 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-32280063

RESUMEN

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.


Asunto(s)
Matriz Extracelular/metabolismo , Lamina Tipo A/metabolismo , Metaloproteinasa 14 de la Matriz/fisiología , Cresta Neural/metabolismo , Animales , Animales Modificados Genéticamente , Cadherinas/metabolismo , Catálisis , Células Cultivadas , Embrión de Pollo , Transición Epitelial-Mesenquimal/fisiología
5.
Development ; 144(13): 2456-2468, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526750

RESUMEN

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.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Inhibición de Contacto , Locomoción , Cresta Neural/citología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Xenopus laevis/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Inhibición de Contacto/efectos de los fármacos , Locomoción/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología
6.
PLoS Comput Biol ; 15(12): e1007171, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31869321

RESUMEN

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.


Asunto(s)
Núcleo Celular/fisiología , Epitelio/embriología , Animales , Tipificación del Cuerpo/fisiología , Recuento de Células , Ciclo Celular/fisiología , Polaridad Celular/fisiología , Proliferación Celular/fisiología , Embrión de Pollo , Biología Computacional , Humanos , Modelos Biológicos , Movimiento/fisiología , Células Neuroepiteliales/citología , Análisis de Sistemas , Anomalías Urogenitales/embriología , Reflujo Vesicoureteral/embriología
7.
PLoS Comput Biol ; 15(4): e1007002, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31009457

RESUMEN

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.


Asunto(s)
Morfogénesis/fisiología , Cresta Neural/embriología , Animales , Anuros/embriología , Adhesión Celular/fisiología , Biología Computacional , Embrión de Mamíferos/embriología , Embrión no Mamífero/embriología , Ratones , Modelos Biológicos , Rombencéfalo/embriología
9.
Genesis ; 56(6-7): e23107, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29675839

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Cresta Neural/embriología , Animales , Adhesión Celular/fisiología , Transición Epitelial-Mesenquimal/fisiología , Humanos
10.
Development ; 140(11): 2247-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23674598

RESUMEN

The neural crest (NC) is a highly migratory multipotent cell population that forms at the interface between the neuroepithelium and the prospective epidermis of a developing embryo. Following extensive migration throughout the embryo, NC cells eventually settle to differentiate into multiple cell types, ranging from neurons and glial cells of the peripheral nervous system to pigment cells, fibroblasts to smooth muscle cells, and odontoblasts to adipocytes. NC cells migrate in large numbers and their migration is regulated by multiple mechanisms, including chemotaxis, contact-inhibition of locomotion and cell sorting. Here, we provide an overview of NC formation, differentiation and migration, highlighting the molecular mechanisms governing NC migration.


Asunto(s)
Movimiento Celular , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/fisiología , Animales , Quimiotaxis , Embrión de Pollo , Desarrollo Embrionario/fisiología , Transición Epitelial-Mesenquimal , Humanos , Ratones , Neoplasias/metabolismo , Cresta Neural/embriología , Xenopus , Pez Cebra
11.
Development ; 140(23): 4763-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24173803

RESUMEN

There is growing evidence that contact inhibition of locomotion (CIL) is essential for morphogenesis and its failure is thought to be responsible for cancer invasion; however, the molecular bases of this phenomenon are poorly understood. Here we investigate the role of the polarity protein Par3 in CIL during migration of the neural crest, a highly migratory mesenchymal cell type. In epithelial cells, Par3 is localised to the cell-cell adhesion complex and is important in the definition of apicobasal polarity, but the localisation and function of Par3 in mesenchymal cells are not well characterised. We show in Xenopus and zebrafish that Par3 is localised to the cell-cell contact in neural crest cells and is essential for CIL. We demonstrate that the dynamics of microtubules are different in different parts of the cell, with an increase in microtubule catastrophe at the collision site during CIL. Par3 loss-of-function affects neural crest migration by reducing microtubule catastrophe at the site of cell-cell contact and abrogating CIL. Furthermore, Par3 promotes microtubule catastrophe by inhibiting the Rac-GEF Trio, as double inhibition of Par3 and Trio restores microtubule catastrophe at the cell contact and rescues CIL and neural crest migration. Our results demonstrate a novel role of Par3 during neural crest migration, which is likely to be conserved in other processes that involve CIL such as cancer invasion or cell dispersion.


Asunto(s)
Proteínas Portadoras/fisiología , Moléculas de Adhesión Celular/metabolismo , Inhibición de Contacto , Microtúbulos/metabolismo , Cresta Neural/embriología , Proteínas de Xenopus/fisiología , Xenopus laevis/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Proteínas Portadoras/genética , Adhesión Celular , Movimiento Celular , Células Cultivadas , Morfogénesis , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/metabolismo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
12.
Biochem J ; 457(1): 19-26, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24325550

RESUMEN

The neural crest is an embryonic stem cell population whose migratory behaviour has been likened to malignant invasion. The neural crest, as does cancer, undergoes an epithelial-to-mesenchymal transition and migrates to colonize almost all the tissues of the embryo. Neural crest cells exhibit collective cell migration, moving in streams of high directionality. The migratory neural crest streams are kept in shape by the presence of negative signals in their vicinity. The directionality of the migrating neural crest is achieved by contact-dependent cell polarization, in a phenomenon called contact inhibition of locomotion. Two cells experiencing contact inhibition of locomotion move away from each other after collision. However, if the cell density is high only cells exposed to a free edge can migrate away from the cluster leading to the directional migration of the whole group. Recent work performed in chicks, zebrafish and frogs has shown that the non-canonical Wnt-PCP (planar cell polarity) pathway plays a major role in neural crest migration. PCP signalling controls contact inhibition of locomotion between neural crest cells by localizing different PCP proteins at the site of cell contact during collision and locally regulating the activity of Rho GTPases. Upon collision RhoA (ras homologue family member A) is activated, whereas Rac1 is inhibited at the contact between two migrating neural crest cells, leading to the collapse of protrusions and the migration of cells away from one another. The present review summarizes the mechanisms that control neural crest migration and focuses on the role of non-canonical Wnt or PCP signalling in this process.


Asunto(s)
Polaridad Celular/fisiología , Cresta Neural/embriología , Vía de Señalización Wnt/fisiología , Animales , Tipificación del Cuerpo/genética , Movimiento Celular/genética , Humanos , Modelos Biológicos , Cresta Neural/fisiología
13.
Cell Mol Life Sci ; 70(19): 3481-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23314710

RESUMEN

Directional cell migration is required for proper embryogenesis, immunity, and healing, and its underpinning regulatory mechanisms are often hijacked during diseases such as chronic inflammations and cancer metastasis. Studies on migratory epithelial tissues have revealed that cells can move as a collective group with shared responsibilities. First thought to be restricted to proper epithelial cell types able to maintain stable cell-cell junctions, the field of collective cell migration is now widening to include cooperative behavior of mesenchymal cells. In this review, we give an overview of the mechanisms driving collective cell migration in epithelial tissues and discuss how mesenchymal cells can cooperate to behave as a collective in the absence of bona fide cell-cell adhesions.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Células Epiteliales/fisiología , Células Madre Mesenquimatosas/citología , Animales , Adhesión Celular/fisiología
14.
Med Sci (Paris) ; 30(8-9): 751-7, 2014.
Artículo en Francés | MEDLINE | ID: mdl-25174751

RESUMEN

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.


Asunto(s)
Movimiento Celular/fisiología , Animales , Comunicación Celular/fisiología , Polaridad Celular/fisiología , Desarrollo Embrionario/fisiología , Humanos , Inmunidad/fisiología , Transducción de Señal , Cicatrización de Heridas/fisiología
15.
Dev Biol ; 366(1): 34-54, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22261150

RESUMEN

After induction and specification in the ectoderm, at the border of the neural plate, the neural crest (NC) population leaves its original territory through a delamination process. Soon afterwards, the NC cells migrate throughout the embryo and colonize a myriad of tissues and organs where they settle and differentiate. The delamination involves a partial or complete epithelium-to-mesenchyme transition (EMT) regulated by a complex network of transcription factors including several proto-oncogenes. Studying the relationship between these genes at the time of emigration, and their individual or collective impact on cell behavior, provides valuable information about their role in EMT in other contexts such as cancer metastasis. During migration, NC cells are exposed to large number of positive and negative regulators that control where they go by generating permissive and restricted areas and by modulating their motility and directionality. In addition, as most NC cells migrate collectively, cell-cell interactions play a crucial role in polarizing the cells and interpreting external cues. Cell cooperation eventually generates an overall polarity to the population, leading to directional collective cell migration. This review will summarize our current knowledge on delamination, EMT and migration of NC cells using key examples from chicken, Xenopus, zebrafish and mouse embryos. Given the similarities between neural crest migration and cancer invasion, these cells may represent a useful model for understanding the mechanisms of metastasis.


Asunto(s)
Movimiento Celular , Transición Epitelial-Mesenquimal , Regulación del Desarrollo de la Expresión Génica , Cresta Neural , Animales , Comunicación Celular , Humanos , Ratones , Invasividad Neoplásica , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/fisiología , Xenopus , Pez Cebra
16.
Genesis ; 49(4): 164-76, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21157935

RESUMEN

The cephalic neural crest (NC) cells delaminate from the neuroepithelium in large numbers and undergo collective cell migration under the influence of multiple factors including positive and negative taxis, cell-cell interactions mediating cell sorting, cell cooperation, and Contact-Inhibition of Locomotion. The migration has to be tightly regulated to allow NC cells to reach precise locations in order to contribute to various craniofacial structures such as the skeletal and peripheral nervous systems. Several birth defects, syndromes, and malformations are due to improper cephalic NC (CNC) migration, and NC cell migration bears important similarities to cancer cell invasion and metastasis dissemination. Therefore, understanding how CNC cells interpret multiple inputs to achieve directional collective cell migration will shed light on pathological situations where cell migration is involved.


Asunto(s)
Movimiento Celular/fisiología , Desarrollo Embrionario/fisiología , Desarrollo Maxilofacial/fisiología , Cresta Neural/fisiología , Cráneo/embriología , Animales , Huesos/embriología , Comunicación Celular/fisiología , Humanos , Metaloproteasas/metabolismo , Sistema Nervioso Periférico/embriología , Cráneo/anatomía & histología
18.
Methods Mol Biol ; 2179: 257-274, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32939726

RESUMEN

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.


Asunto(s)
Rastreo Celular/métodos , Transición Epitelial-Mesenquimal , Cresta Neural/citología , Cultivo Primario de Células/métodos , Animales , Adhesión Celular , Movimiento Celular , Rastreo Celular/instrumentación , Xenopus
19.
Cells Dev ; 168: 203723, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34284169

RESUMEN

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.


Asunto(s)
Pollos , Tubo Neural , Animales , División Celular , Sistema Nervioso Central , Mesodermo , Tubo Neural/metabolismo
20.
Front Physiol ; 11: 586432, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324240

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA