Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
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
2.
Semin Cell Dev Biol ; 147: 12-23, 2023 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-37002130

RESUMEN

The process by which biological systems such as cells, tissues and organisms acquire shape has been named as morphogenesis and it is central to a plethora of biological contexts including embryo development, wound healing, or even cancer. Morphogenesis relies in both self-organising properties of the system and in environmental inputs (biochemical and biophysical). The classical view of morphogenesis is based on the study of external biochemical molecules, such as morphogens. However, recent studies are establishing that the mechanical environment is also used by cells to communicate within tissues, suggesting that this mechanical crosstalk is essential to synchronise morphogenetic transitions and self-organisation. In this article we discuss how tissue interaction drive robust morphogenesis, starting from a classical biochemical view, to finalise with more recent advances on how the biophysical properties of a tissue feedback with their surroundings to allow form acquisition. We also comment on how in silico models aid to integrate and predict changes in cell and tissue behaviour. Finally, considering recent advances from the developmental biomechanics field showing that mechanical inputs work as cues that promote morphogenesis, we invite to revisit the concept of morphogen.


Asunto(s)
Desarrollo Embrionario , Transducción de Señal , Morfogénesis , Fenómenos Biomecánicos , Modelos Biológicos
3.
J Cell Sci ; 136(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37747423

RESUMEN

Shear stress is essential for normal physiology and malignancy. Common physiological processes - such as blood flow, particle flow in the gut, or contact between migratory cell clusters and their substrate - produce shear stress that can have an impact on the behavior of different tissues. In addition, shear stress has roles in processes of biomedical interest, such as wound healing, cancer and fibrosis induced by soft implants. Thus, understanding how cells react and adapt to shear stress is important. In this Review, we discuss in vivo and in vitro data obtained from vascular and epithelial models; highlight the insights these have afforded regarding the general mechanisms through which cells sense, transduce and respond to shear stress at the cellular levels; and outline how the changes cells experience in response to shear stress impact tissue organization. Finally, we discuss the role of shear stress in collective cell migration, which is only starting to be appreciated. We review our current understanding of the effects of shear stress in the context of embryo development, cancer and fibrosis, and invite the scientific community to further investigate the role of shear stress in these scenarios.


Asunto(s)
Desarrollo Embrionario , Cicatrización de Heridas , Movimiento Celular , Estrés Mecánico
4.
Nature ; 554(7693): 523-527, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29443958

RESUMEN

Collective cell migration is essential for morphogenesis, tissue remodelling and cancer invasion. In vivo, groups of cells move in an orchestrated way through tissues. This movement involves mechanical as well as molecular interactions between cells and their environment. While the role of molecular signals in collective cell migration is comparatively well understood, how tissue mechanics influence collective cell migration in vivo remains unknown. Here we investigated the importance of mechanical cues in the collective migration of the Xenopus laevis neural crest cells, an embryonic cell population whose migratory behaviour has been likened to cancer invasion. We found that, during morphogenesis, the head mesoderm underlying the cephalic neural crest stiffens. This stiffening initiates an epithelial-to-mesenchymal transition in neural crest cells and triggers their collective migration. To detect changes in their mechanical environment, neural crest cells use mechanosensation mediated by the integrin-vinculin-talin complex. By performing mechanical and molecular manipulations, we show that mesoderm stiffening is necessary and sufficient to trigger neural crest migration. Finally, we demonstrate that convergent extension of the mesoderm, which starts during gastrulation, leads to increased mesoderm stiffness by increasing the cell density underneath the neural crest. These results show that convergent extension of the mesoderm has a role as a mechanical coordinator of morphogenesis, and reveal a link between two apparently unconnected processes-gastrulation and neural crest migration-via changes in tissue mechanics. Overall, we demonstrate that changes in substrate stiffness can trigger collective cell migration by promoting epithelial-to-mesenchymal transition in vivo. More broadly, our results raise the idea that tissue mechanics combines with molecular effectors to coordinate morphogenesis.


Asunto(s)
Movimiento Celular , Mecanotransducción Celular , Mesodermo/fisiología , Morfogénesis , Cresta Neural/citología , Xenopus laevis/embriología , Animales , Transición Epitelial-Mesenquimal , Matriz Extracelular , Femenino , Gastrulación , Dureza , Integrinas/metabolismo , Mesodermo/citología , Mesodermo/embriología
5.
Dev Biol ; 492: 79-86, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36206829

RESUMEN

Collective cell migration is essential for embryonic development, tissue regeneration and repair, and has been implicated in pathological conditions such as cancer metastasis. It is, in part, directed by external cues that promote front-to-rear polarity in individual cells. However, our understanding of the pathways that underpin the directional movement of cells in response to external cues remains incomplete. To examine this issue we made use of neural crest cells (NC), which migrate as a collective during development to generate vital structures including bones and cartilage. Using a candidate approach, we found an essential role for Ran-binding protein 1 (RanBP1), a key effector of the nucleocytoplasmic transport pathway, in enabling directed migration of these cells. Our results indicate that RanBP1 is required for establishing front-to-rear polarity, so that NCs are able to chemotax. Moreover, our work suggests that RanBP1 function in chemotaxis involves the polarity kinase LKB1/PAR4. We envisage that regulated nuclear export of LKB1 through Ran/RanBP1 is a key regulatory step required for establishing front-to-rear polarity and thus chemotaxis, during NC collective migration.


Asunto(s)
Cresta Neural , Proteínas Nucleares , Embarazo , Femenino , Humanos , Cresta Neural/metabolismo , Proteínas Nucleares/metabolismo , Movimiento Celular/fisiología , Quimiotaxis
6.
Nat Mater ; 21(11): 1314-1323, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35970965

RESUMEN

Embryogenesis, tissue repair and cancer metastasis rely on collective cell migration. In vitro studies propose that cells are stiffer while migrating in stiff substrates, but softer when plated in compliant surfaces which are typically considered as non-permissive for migration. Here we show that cells within clusters from embryonic tissue dynamically decrease their stiffness in response to the temporal stiffening of their native substrate to initiate collective cell migration. Molecular and mechanical perturbations of embryonic tissues reveal that this unexpected mechanical response involves a mechanosensitive pathway relying on Piezo1-mediated microtubule deacetylation. We further show that decreasing microtubule acetylation and consequently cluster stiffness is sufficient to trigger collective cell migration in soft non-permissive substrates. This suggests that reaching an optimal cluster-to-substrate stiffness ratio is essential to trigger the onset of this collective process. Overall, these in vivo findings challenge the current understanding of collective cell migration and its physiological and pathological roles.


Asunto(s)
Movimiento Celular
7.
Semin Cell Dev Biol ; 93: 55-68, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-29859995

RESUMEN

Cell migration is essential for a wide range of biological processes such as embryo morphogenesis, wound healing, regeneration, and also in pathological conditions, such as cancer. In such contexts, cells are required to migrate as individual entities or as highly coordinated collectives, both of which requiring cells to respond to molecular and mechanical cues from their environment. However, whilst the function of chemical cues in cell migration is comparatively well understood, the role of tissue mechanics on cell migration is just starting to be studied. Recent studies suggest that the dynamic tuning of the viscoelasticity within a migratory cluster of cells, and the adequate elastic properties of its surrounding tissues, are essential to allow efficient collective cell migration in vivo. In this review we focus on the role of viscoelasticity in the control of collective cell migration in various cellular systems, mentioning briefly some aspects of single cell migration. We aim to provide details on how viscoelasticity of collectively migrating groups of cells and their surroundings is adjusted to ensure correct morphogenesis, wound healing, and metastasis. Finally, we attempt to show that environmental viscoelasticity triggers molecular changes within migrating clusters and that these new molecular setups modify clusters' viscoelasticity, ultimately allowing them to migrate across the challenging geometries of their microenvironment.


Asunto(s)
Movimiento Celular , Humanos , Termodinámica , Viscosidad
8.
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
9.
Dev Biol ; 444 Suppl 1: S202-S208, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29522707

RESUMEN

The neural crest is a multipotent population of cells that originates a variety of cell types. Many animal models are used to study neural crest induction, migration and differentiation, with amphibians and birds being the most widely used systems. A major technological advance to study neural crest development in mouse, chick and zebrafish has been the generation of transgenic animals in which neural crest specific enhancers/promoters drive the expression of either fluorescent proteins for use as lineage tracers, or modified genes for use in functional studies. Unfortunately, no such transgenic animals currently exist for the amphibians Xenopus laevis and tropicalis, key model systems for studying neural crest development. Here we describe the generation and characterization of two transgenic Xenopus laevis lines, Pax3-GFP and Sox10-GFP, in which GFP is expressed in the pre-migratory and migratory neural crest, respectively. We show that Pax3-GFP could be a powerful tool to study neural crest induction, whereas Sox10-GFP could be used in the study of neural crest migration in living embryos.


Asunto(s)
Cresta Neural/metabolismo , Factor de Transcripción PAX3/metabolismo , Factores de Transcripción SOXE/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica/genética , Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes , Humanos , Cresta Neural/embriología , Cresta Neural/fisiología , Neurogénesis , Factor de Transcripción PAX3/fisiología , Factores de Transcripción SOXE/fisiología , Xenopus laevis/embriología
10.
Development ; 143(12): 2194-205, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27122165

RESUMEN

Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/ß-catenin signalling. Using two in vivo models, we show that Wnt/ß-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/ß-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of ß-catenin, preventing ß-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits ß-catenin signalling, which then affects NC delamination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Wnt/metabolismo , Animales , Movimiento Celular , Núcleo Celular/metabolismo , Embrión de Pollo , Femenino , Células HEK293 , Humanos , Fracciones Subcelulares/metabolismo , Vía de Señalización Wnt , Xenopus laevis/embriología , Xenopus laevis/metabolismo , beta Catenina/metabolismo
11.
Development ; 142(9): 1555-60, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25922521

RESUMEN

The neural crest is a uniquely vertebrate cell type and has been well studied in a number of model systems. Zebrafish, Xenopus and chick embryos largely show consistent requirements for specific genes in early steps of neural crest development. By contrast, knockouts of homologous genes in the mouse often do not exhibit comparable early neural crest phenotypes. In this Spotlight article, we discuss these species-specific differences, suggest possible explanations for the divergent phenotypes in mouse and urge the community to consider these issues and the need for further research in complementary systems.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes/métodos , Modelos Animales , Cresta Neural/embriología , Fenotipo , Animales , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Especificidad de la Especie , Xenopus , Pez Cebra
12.
FEBS J ; 289(10): 2736-2754, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33811732

RESUMEN

Directed cell migration is essential for cells to efficiently migrate in physiological and pathological processes. While migrating in their native environment, cells interact with multiple types of cues, such as mechanical and chemical signals. The role of chemical guidance via chemotaxis has been studied in the past, the understanding of mechanical guidance of cell migration via durotaxis remained unclear until very recently. Nonetheless, durotaxis has become a topic of intensive research and several advances have been made in the study of mechanically guided cell migration across multiple fields. Thus, in this article we provide a state of the art about durotaxis by discussing in silico, in vitro and in vivo data. We also present insights on the general mechanisms by which cells sense, transduce and respond to environmental mechanics, to then contextualize these mechanisms in the process of durotaxis and explain how cells bias their migration in anisotropic substrates. Furthermore, we discuss what is known about durotaxis in vivo and we comment on how haptotaxis could arise from integrating durotaxis and chemotaxis in native environments.


Asunto(s)
Quimiotaxis , Movimiento Celular/fisiología
13.
Methods Mol Biol ; 2438: 495-515, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35147960

RESUMEN

During vertebrate embryogenesis, tissues interact and influence each other's development to shape an embryo. While communication by molecular components has been extensively explored, the role of mechanical interaction between tissues during embryogenesis is just starting to be revealed. Addressing mechanical involvement in morphogenesis has traditionally been challenging mainly due to the lack of proper tools to measure and modify mechanical environments of cells in vivo. We have recently used atomic force microscopy (AFM) to show that the migration of the Xenopus laevis cephalic neural crest cells is triggered by stiffening of the mesoderm, a tissue that neural crest cells use as a migratory substrate in vivo. Interestingly we showed that the activity of the planar cell polarity (PCP) pathway is required to mediate this novel mechanical interaction between two tissues. In this chapter, we share the toolbox that we developed to study the role of PCP signaling in mesoderm cell accumulation and stiffening (in vivo) as well as the impact of mesoderm stiffness in promoting neural crest cell polarity and migration (ex vivo). We believe that these tools can be of general use for investigators interested in addressing the role of mechanical inputs in vivo and ex vivo.


Asunto(s)
Polaridad Celular , Cresta Neural , Animales , Movimiento Celular , Mesodermo , Cresta Neural/metabolismo , Xenopus laevis
14.
BMC Evol Biol ; 10: 78, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20236534

RESUMEN

BACKGROUND: The mineralized skeleton is a major evolutionary novelty that has contributed to the impressive morphological diversifications of the vertebrates. Essential to bone biology is the solidified extracellular matrix secreted by highly specialized cells, the osteoblasts. We now have a rather complete view of the events underlying osteogenesis, from a cellular, molecular, genetic, and epigenetic perspective. Because this knowledge is still largely restricted to mammals, it is difficult, if not impossible, to deduce the evolutionary history of the regulatory network involved in osteoblasts specification and differentiation. In this study, we focused on the transcriptional regulators Runx2 and VDR (the Vitamin D Receptor) that, in mammals, directly interact together and stabilize complexes of co-activators and chromatin remodellers, thereby allowing the transcriptional activation of target genes involved in extracellular matrix mineralization. Using a combination of functional, biochemical, and histological approaches, we have asked if the interaction observed between Runx2 and VDR represents a recent mammalian innovation, or if it results from more ancient changes that have occurred deep in the vertebrate lineage. RESULTS: Using immunohistochemistry and in situ hybridization in developing embryos of chick, frog and teleost fishes, we have revealed that the co-expression of Runx2 and VDR in skeletal elements has been particularly strengthened in the lineage leading to amniotes. We show that the teleost Runx2 orthologue as well as the three mammalian Runx1, Runx2 and Runx3 paralogues are able to co-immunoprecipitate with the VDR protein present in nuclear extracts of rat osteoblasts stimulated with 1alpha,25-dihydroxyvitamin D3. In addition, the teleost Runx2 can activate the transcription of the mammalian osteocalcin promoter in transfection experiments, and this response can be further enhanced by 1alpha,25-dihydroxyvitamin D3. Finally, using pull-down experiments between recombinant proteins, we show that the VDR homologue from teleosts, but not from ascidians, is able to directly interact with the mammalian Runx2 homologue. CONCLUSIONS: We propose an evolutionary scenario for the assembly of the molecular machinery involving Runx2 and VDR in vertebrates. In the last common ancestor of actinopterygians and sacropterygians, the three Runx paralogues possessed the potential to physically and functionally interact with the VDR protein. Therefore, 1alpha,25-dihydroxyvitamin D3 might have been able to modulate the transcriptional activity of Runx1, Runx2 or Runx3 in the tissues expressing VDR. After the split from amphibians, in the lineage leading to amniotes, Runx2 and VDR became robustly co-expressed in developing skeletal elements, and their regulatory interaction was incorporated in the genetic program involved in the specification and differentiation of osteoblasts.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Evolución Molecular , Osteogénesis , Receptores de Calcitriol/genética , Vertebrados/genética , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/citología , Receptores de Calcitriol/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vertebrados/embriología , Vertebrados/metabolismo
15.
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.

16.
Nat Cell Biol ; 22(7): 803-814, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32572169

RESUMEN

Cell shape is controlled by the submembranous cortex, an actomyosin network mainly generated by two actin nucleators: the Arp2/3 complex and the formin mDia1. Changes in relative nucleator activity may alter cortical organization, mechanics and cell shape. Here we investigate how nucleation-promoting factors mediate interactions between nucleators. In vitro, the nucleation-promoting factor SPIN90 promotes formation of unbranched filaments by Arp2/3, a process thought to provide the initial filament for generation of dendritic networks. Paradoxically, in cells, SPIN90 appears to favour a formin-dominated cortex. Our in vitro experiments reveal that this feature stems mainly from two mechanisms: efficient recruitment of mDia1 to SPIN90-Arp2/3 nucleated filaments and formation of a ternary SPIN90-Arp2/3-mDia1 complex that greatly enhances filament nucleation. Both mechanisms yield rapidly elongating filaments with mDia1 at their barbed ends and SPIN90-Arp2/3 at their pointed ends. Thus, in networks, SPIN90 lowers branching densities and increases the proportion of long filaments elongated by mDia1.


Asunto(s)
Citoesqueleto de Actina/fisiología , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Forminas/metabolismo , Melanoma/patología , Proteínas Musculares/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Blástula/citología , Blástula/metabolismo , Forma de la Célula , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Forminas/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteínas Musculares/genética , Xenopus laevis/crecimiento & desarrollo , Xenopus laevis/metabolismo
17.
Methods Mol Biol ; 1976: 135-152, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30977071

RESUMEN

The neural crest is an embryonic cell population induced at the border of the neural plate from where it delaminates and migrates long distances across the embryo. Due to its extraordinary migratory capabilities, the neural crest has become a powerful system to study cellular and molecular aspects of collective and single cell migration both in vivo and in vitro. Here we provide detailed protocols used to perform quantitative analysis of molecular and cellular aspects of Xenopus laevis neural crest cell migration, both in vivo and in vitro.


Asunto(s)
Movimiento Celular/fisiología , Cresta Neural/citología , Animales , Adhesión Celular/fisiología , Movimiento Celular/genética , Placa Neural/citología , Transducción de Señal , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
18.
Nat Commun ; 9(1): 3846, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242148

RESUMEN

Connexins are the primary components of gap junctions, providing direct links between cells under many physiological processes. Here, we demonstrate that in addition to this canonical role, Connexins act as transcriptional regulators. We show that Connexin 43 (Cx43) controls neural crest cell migration in vivo by directly regulating N-cadherin transcription. This activity requires interaction between Cx43 carboxy tail and the basic transcription factor-3, which drives the translocation of Cx43 tail to the nucleus. Once in the nucleus they form a complex with PolII which directly binds to the N-cadherin promoter. We found that this mechanism is conserved between amphibian and mammalian cells. Given the strong evolutionary conservation of connexins across vertebrates, this may reflect a common mechanism of gene regulation by a protein whose function was previously ascribed only to gap junctional communication.


Asunto(s)
Cadherinas/metabolismo , Conexina 43/metabolismo , Regulación de la Expresión Génica , Cresta Neural/fisiología , Animales , Movimiento Celular , ADN Polimerasa II/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Xenopus laevis
19.
Nat Commun ; 9(1): 4633, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30385761

RESUMEN

The original version of this Article contained an error in the spelling of the author Alexandra Schambony, which was incorrectly given as Alexandra Schambon. This has now been corrected in both the PDF and HTML versions of the Article.

20.
Curr Top Dev Biol ; 112: 301-23, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25733144

RESUMEN

Cell migration is essential for morphogenesis, adult tissue remodeling, wound healing, and cancer cell migration. Cells can migrate as individuals or groups. When cells migrate in groups, cell-cell interactions are crucial in order to promote the coordinated behavior, essential for collective migration. Interestingly, recent evidence has shown that cell-cell interactions are also important for establishing and maintaining the directionality of these migratory events. We focus on neural crest cells, as they possess extraordinary migratory capabilities that allow them to migrate and colonize tissues all over the embryo. Neural crest cells undergo an epithelial-to-mesenchymal transition at the same time than perform directional collective migration. Cell-cell adhesion has been shown to be an important source of planar cell polarity and cell coordination during collective movement. We also review molecular mechanisms underlying cadherin turnover, showing how the modulation and dynamics of cell-cell adhesions are crucial in order to maintain tissue integrity and collective migration in vivo. We conclude that cell-cell adhesion during embryo development cannot be considered as simple passive resistance to force, but rather participates in signaling events that determine important cell behaviors required for cell migration.


Asunto(s)
Adhesión Celular/fisiología , Comunicación Celular , Movimiento Celular/fisiología , Sistema Nervioso/embriología , Cresta Neural/citología , Animales , Cadherinas/metabolismo , Desarrollo Embrionario , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA