首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 10 毫秒
1.
2.
During vertebrate gastrulation, the three germ layers, ectoderm, mesoderm and endoderm are formed, and the resulting progenitor cells are brought into the positions from which they will later contribute more complex tissues and organs. A core element in this process is the internalization of mesodermal and endodermal progenitors at the onset of gastrulation. Although many of the molecules that induce mesendoderm have been identified, much less is known about the cellular mechanisms underlying mesendodermal cell internalization and germ layer formation. Here we show that at the onset of zebrafish gastrulation, mesendodermal progenitors in dorsal/axial regions of the germ ring internalize by single cell delamination. Once internalized, mesendodermal progenitors upregulate E-Cadherin (Cadherin 1) expression, become increasingly motile and eventually migrate along the overlying epiblast (ectodermal) cell layer towards the animal pole of the gastrula. When E-Cadherin function is compromised, mesendodermal progenitors still internalize, but, with gastrulation proceeding, fail to elongate and efficiently migrate along the epiblast, whereas epiblast cells themselves exhibit reduced radial cell intercalation movements. This indicates that cadherin-mediated cell-cell adhesion is needed within the forming shield for both epiblast cell intercalation, and mesendodermal progenitor cell elongation and migration during zebrafish gastrulation. Our data provide insight into the cellular mechanisms underlying mesendodermal progenitor cell internalization and subsequent migration during zebrafish gastrulation, and the role of cadherin-mediated cell-cell adhesion in these processes.  相似文献   

3.
Knowledge of the molecular mechanisms regulating cell ingression, epithelial–mesenchymal transition and migration movements during amniote gastrulation is steadily improving. In the frog and fish embryo, Wnt5 and Wnt11 ligands are expressed around the blastopore and play an important role in regulating cell movements associated with gastrulation. In the chicken embryo, although Wnt5a and Wnt5b are expressed in the primitive streak, the known Wnt11 gene is expressed in paraxial and intermediate mesoderm, and in differentiated myocardial cells, but not in the streak. Here, we identify a previously uncharacterized chicken Wnt11 gene, Wnt11b, that is orthologous to the frog Wnt11 and zebrafish Wnt11 (silberblick) genes. Chicken Wnt11b is expressed in the primitive streak in a pattern similar to chicken Wnt5a and Wnt5b. When non-canonical Wnt signaling is blocked using a Dishevelled dominant-negative protein, gastrulation movements are inhibited and cells accumulate in the primitive streak. Furthermore, disruption of non-canonical Wnt signaling by overexpression of full-length or dominant-negative Wnt11b or Wnt5a constructions abrogates normal cell migration through the primitive streak. We conclude that non-canonical Wnt signaling, mediated in part by Wnt11b, is important for regulation of gastrulation cell movements in the avian embryo.  相似文献   

4.
5.
During vertebrate gastrulation, both mesodermal and endodermal cells internalize through the blastopore beneath the ectoderm. In zebrafish, the internalized mesodermal cells move towards the dorsal side of the gastrula and, at the same time, they extend anteriorly by convergence and extension (C&E) movements. Endodermal cells showing characteristic filopodia then migrate into the inner layer within the hypoblast next to the yolk syncytial layer (YSL). However, little is known about how the movement of endodermal cells is regulated during gastrulation. Here we show that sdf1a- and sdf1b-expressing mesodermal cells control the movements of the cxcr4a-expressing endodermal cells. The directional migration of endodermal cells during gastrulation is inhibited by knockdown of either cxcr4a or sdf1a/sdf1b (sdf1). We also show that misexpressed Sdf1 acts as a chemoattractant for cxcr4a-expressing endodermal cells. We further found, using the endoderm-specific transgenic line Tg(sox17:EGFP), that Sdf1/Cxcr4 signaling regulates both the formation and orientation of filopodial processes in endodermal cells. Moreover, the accumulation of phosphoinositide 3,4,5-trisphosphate (PIP(3)), which is known to occur at the leading edge of migrating cells, is not observed at the filopodia of endodermal cells. Based on our results, we propose that sdf1-expressing mesodermal cells, which overlie the endodermal layer, guide the cxcr4a-expressing endodermal cells to the dorsal side of the embryo during gastrulation, possibly through a PIP(3)-independent pathway.  相似文献   

6.
Wnt genes play important roles in regulating patterning and morphogenesis during vertebrate gastrulation. In zebrafish, slb/wnt11 is required for convergence and extension movements, but not cell fate specification during gastrulation. To determine if other Wnt genes functionally interact with slb/wnt11, we analysed the role of ppt/wnt5 during zebrafish gastrulation. ppt/wnt5 is maternally provided and zygotically expressed at all stages during gastrulation. The analysis of ppt mutant embryos reveals that Ppt/Wnt5 regulates cell elongation and convergent extension movements in posterior regions of the gastrula, while its function in more anterior regions is largely redundant to that of Slb/Wnt11. Frizzled-2 functions downstream of ppt/wnt5, indicating that it might act as a receptor for Ppt/Wnt5 in this process. The characterisation of the role of Ppt/Wnt5 provides insight into the functional diversity of Wnt genes in regulating vertebrate gastrulation movements.  相似文献   

7.
Key to invasiveness is the ability of tumor cells to modify the extracellular matrix, become motile, and engage in directed migration towards the vasculature. One significant protein associated with metastatic progression is membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP14). How MMP14 activity is coordinated with other signaling pathways to regulate cell migration in vivo is largely unknown. Here we have used zebrafish embryogenesis as a model to understand the potential relationship between MMP14-dependent pericellular proteolysis, cell polarity, and motility. Knockdown of zebrafish Mmp14 function disrupted gastrulation convergence and extension cell movements and craniofacial morphogenesis. Using time-lapse imaging and morphometric analyses, we show that Mmp14 is required for proper cell polarity underlying the directed migration of mesodermal cells during gastrulation. We have identified a genetic interaction between mmp14 and non-canonical Wnt signaling, a pathway that also regulates cell polarity in embryonic tissues and is increasingly being linked with tumor cell migration. Finally, we demonstrate that Van Gogh-like 2, a key regulator of the non-canonical Wnt pathway, co-localizes with MMP14 and becomes redistributed towards the leading edge of polarized human cancer cells. Together, our results support the notion that pathways regulating pericellular proteolysis and cell polarity converge to promote efficient cell migration.  相似文献   

8.
We have reviewed the evidence supporting the notion that the fibrillar extracellular matrix on the basal surface of the blastocoel roof in amphibian embryos directs and guides mesodermal cell migration during gastrulation. Based on extensive experimental evidence in several different systems, we conclude the following: (i) the fibrillar extracellular matrix contains fibronectin (FN) and laminin. (ii) The fibrils are oriented in such a way as to promote directional migration of mesodermal cells during migration. (iii) We have used several different probes to disrupt the interaction between migrating mesodermal cells and the fibrillar extracellular matrix. These probes include: (a) nucleocytoplasmic and interspecific hybridization. Such embryos have defects in FN synthesis and gastrulation. (b) Fab' fragments of anti-FN and anti-integrin VLA-5 IgGs prohibit mesodermal cell adhesion both in vitro and in vivo and gastrulation is arrested. (c) Peptides containing the RGDS sequence specifically inhibit interactions between migrating mesodermal cells and the FN-fibrillar matrix. (d) Tenascin blocks cell adhesion to FN in vitro and gastrulation in vivo. (e) Antibodies against the cytoplasmic domain of beta 1 integrin, when injected into blastomeres, prevent FN-fibrillogenesis in progeny of injected blastomeres and delay mesodermal cell migration selectively in the progeny of injected blastomeres but not in the uninjected blastomere progeny.  相似文献   

9.
During vertebrate gastrulation, mesodermal and ectodermal cells undergo convergent extension, a process characterised by prominent cellular rearrangements in which polarised cells intercalate along the medio-lateral axis leading to elongation of the antero-posterior axis. Recently, it has become evident that a noncanonical Wnt/Frizzled (Fz)/Dishevelled (Dsh) signalling pathway, which is related to the planar-cell-polarity (PCP) pathway in flies, regulates convergent extension during vertebrate gastrulation. Here we isolate and functionally characterise a zebrafish homologue of Drosophila prickle (pk), a gene that is implicated in the regulation of PCP. Zebrafish pk1 is expressed maternally and in moving mesodermal precursors. Abrogation of Pk1 function by morpholino oligonucleotides leads to defective convergent extension movements, enhances the silberblick (slb)/wnt11 and pipetail (Ppt)/wnt5 phenotypes and suppresses the ability of Wnt11 to rescue the slb phenotype. Gain-of-function of Pk1 also inhibits convergent extension movements and enhances the slb phenotype, most likely caused by the ability of Pk1 to block the Fz7-dependent membrane localisation of Dsh by downregulating levels of Dsh protein. Furthermore, we show that pk1 interacts genetically with trilobite (tri)/strabismus to mediate the caudally directed migration of cranial motor neurons and convergent extension. These results indicate that, during zebrafish gastrulation Pk1 acts, in part, through interaction with the noncanonical Wnt11/Wnt5 pathway to regulate convergent extension cell movements, but is unlikely to simply be a linear component of this pathway. In addition, Pk1 interacts with Tri to mediate posterior migration of branchiomotor neurons, probably independent of the noncanonical Wnt pathway.  相似文献   

10.
11.
Mutations in the zebrafish knypek locus impair gastrulation movements of convergent extension that narrow embryonic body and elongate it from head to tail. We demonstrate that knypek regulates cellular movements but not cell fate specification. Convergent extension movement defects in knypek are associated with abnormal cell polarity, as mutant cells fail to elongate and align medio-laterally. Positional cloning reveals that knypek encodes a member of the glypican family of heparan sulfate proteoglycans. Double mutant and overexpression analyses show that Knypek potentiates Wnt11 signaling, mediating convergent extension. These studies provide experimental and genetic evidence that glypican Knypek acts during vertebrate gastrulation as a positive modulator of noncanonical Wnt signaling to establish polarized cell behaviors underlying convergent extension movements.  相似文献   

12.
BACKGROUND: Embryonic patterning mechanisms regulate the cytoskeletal machinery that drives morphogenesis, but there are few cases where links between patterning mechanisms and morphogenesis are well understood. We have used a combination of genetics, in vivo imaging, and cell manipulations to identify such links in C. elegans gastrulation. Gastrulation in C. elegans begins with the internalization of endodermal precursor cells in a process that depends on apical constriction of ingressing cells. RESULTS: We show that ingression of the endodermal precursor cells is regulated by pathways, including a Wnt-Frizzled signaling pathway, that specify endodermal cell fate. We find that Wnt signaling has a role in gastrulation in addition to its earlier roles in regulating endodermal cell fate and cell-cycle timing. In the absence of Wnt signaling, endodermal precursor cells polarize and enrich myosin II apically but fail to contract their apical surfaces. We show that a regulatory myosin light chain normally becomes phosphorylated on the apical side of ingressing cells at a conserved site that can lead to myosin-filament formation and contraction of actomyosin networks and that this phosphorylation depends on Wnt signaling. CONCLUSIONS: We conclude that Wnt signaling regulates C. elegans gastrulation through regulatory myosin light-chain phosphorylation, which results in the contraction of the apical surface of ingressing cells. These findings forge new links between cell-fate specification and morphogenesis, and they represent a novel mechanism by which Wnt signaling can regulate morphogenesis.  相似文献   

13.
Wnt11 is a key signal, determining cell polarization and migration during vertebrate gastrulation. It is known that Wnt11 functionally interacts with several signaling components, the homologues of which control planar cell polarity in Drosophila melanogaster. Although in D. melanogaster these components are thought to polarize cells by asymmetrically localizing at the plasma membrane, it is not yet clear whether their subcellular localization plays a similarly important role in vertebrates. We show that in zebrafish embryonic cells, Wnt11 locally functions at the plasma membrane by accumulating its receptor, Frizzled 7, on adjacent sites of cell contacts. Wnt11-induced Frizzled 7 accumulations recruit the intracellular Wnt signaling mediator Dishevelled, as well as Wnt11 itself, and locally increase cell contact persistence. This increase in cell contact persistence is mediated by the local interaction of Wnt11, Frizzled 7, and the atypical cadherin Flamingo at the plasma membrane, and it does not require the activity of further downstream effectors of Wnt11 signaling, such as RhoA and Rok2. We propose that Wnt11, by interacting with Frizzled 7 and Flamingo, modulates local cell contact persistence to coordinate cell movements during gastrulation.  相似文献   

14.
During vertebrate gastrulation, convergence and extension cell movements are coordinated with the anteroposterior and mediolateral embryonic axes. Wnt planar cell polarity (Wnt/PCP) signaling polarizes the motile behaviors of cells with respect to the anteroposterior embryonic axis. Understanding how Wnt/PCP signaling mediates convergence and extension (C&E) movements requires analysis of the mechanisms employed to alter cell morphology and behavior with respect to embryonic polarity. Here, we examine the interactions between the microtubule cytoskeleton and Wnt/PCP signaling during zebrafish gastrulation. First, we assessed the location of the centrosome/microtubule organizing center (MTOC) relative to the cell nucleus and the body axes, as a marker of cell polarity. The intracellular position of MTOCs was polarized, perpendicular to the plane of the germ layers, independently of Wnt/PCP signaling. In addition, this position became biased posteriorly and medially within the plane of the germ layers at the transition from mid- to late gastrulation and from slow to fast C&E movements. This depends on intact Wnt/PCP signaling through Knypek (Glypican4/6) and Dishevelled components. Second, we tested whether microtubules are required for planar cell polarization. Once the planar cell polarity is established, microtubules are not required for accumulation of Prickle at the anterior cell edge. However, microtubules are needed for cell-cell contacts and initiation of its anterior localization. Reciprocal interactions occur between Wnt/PCP signaling and microtubule cytoskeleton during C&E gastrulation movements. Wnt/PCP signaling influences the polarity of the microtubule cytoskeleton and, conversely, microtubules are required for the asymmetric distribution of Wnt/PCP pathway components.  相似文献   

15.
In Caenorhabditis elegans, Wnt signaling pathways are important in controlling cell polarity and cell migrations. In the embryo, a novel Wnt pathway functions through a (beta)-catenin homolog, WRM-1, to downregulate the levels of POP-1/Tcf in the posterior daughter of the EMS blastomere. The level of POP-1 is also lower in the posterior daughters of many anteroposterior asymmetric cell divisions during development. I have found that this is the case for of a pair of postembryonic blast cells in the tail. In wild-type animals, the level of POP-1 is lower in the posterior daughters of the two T cells, TL and TR. Furthermore, in lin-44/Wnt mutants, in which the polarities of the T cell divisions are frequently reversed, the level of POP-1 is frequently lower in the anterior daughters of the T cells. I have used a novel RNA-mediated interference technique to interfere specifically with pop-1 zygotic function and have determined that pop-1 is required for wild-type T cell polarity. Surprisingly, none of the three C. elegans (beta)-catenin homologs appeared to function with POP-1 to control T cell polarity. Wnt signaling by EGL-20/Wnt controls the migration of the descendants of the QL neuroblast by regulating the expression the Hox gene mab-5. Interfering with pop-1 zygotic function caused defects in the migration of the QL descendants that mimicked the defects in egl-20/Wnt mutants and blocked the expression of mab-5. This suggests that POP-1 functions in the canonical Wnt pathway to control QL descendant migration and in novel Wnt pathways to control EMS and T cell polarities.  相似文献   

16.
The MAPKs are key regulatory signaling molecules in many cellular processes. Here we define differential functions for ERK1 and ERK2 MAPKs in zebrafish embryogenesis. Morpholino knockdown of ERK1 and ERK2 resulted in cell migration defects during gastrulation, which could be rescued by co-injection of the corresponding mRNA. Strikingly, Erk2 mRNA cross-rescued ERK1 knockdown, but erk1 mRNA was unable to compensate for ERK2 knockdown. Cell-tracing experiments revealed a convergence defect for ERK1 morphants without a severe posterior-extension defect, whereas ERK2 morphants showed a more severe reduction in anterior-posterior extension. These defects were primary changes in gastrulation cell movements and not caused by altered cell fate specification. Saturating knockdown conditions showed that the absence of FGF-mediated dual-phosphorylated ERK2 from the blastula margin blocked initiation of epiboly, actin and tubulin cytoskeleton reorganization processes and further arrested embryogenesis, whereas ERK1 knockdown had only a mild effect on epiboly progression. Together, our data define distinct roles for ERK1 and ERK2 in developmental cell migration processes during zebrafish embryogenesis.  相似文献   

17.
Wnt11 plays a central role in tissue morphogenesis during vertebrate gastrulation, but the molecular and cellular mechanisms by which Wnt11 exerts its effects remain poorly understood. Here, we show that Wnt11 functions during zebrafish gastrulation by regulating the cohesion of mesodermal and endodermal (mesendodermal) progenitor cells. Importantly, we demonstrate that Wnt11 activity in this process is mediated by the GTPase Rab5, a key regulator of early endocytosis, as blocking Rab5c activity in wild-type embryos phenocopies slb/wnt11 mutants, and enhancing Rab5c activity in slb/wnt11 mutant embryos rescues the mutant phenotype. In addition, we find that Wnt11 and Rab5c control the endocytosis of E-cadherin and are required in mesendodermal cells for E-cadherin-mediated cell cohesion. Together, our results suggest that Wnt11 controls tissue morphogenesis by modulating E-cadherin-mediated cell cohesion through Rab5c, a novel mechanism of Wnt signaling in gastrulation.  相似文献   

18.
Slow-twitch muscle fibers of the zebrafish myotome undergo a unique set of morphogenetic cell movements. During embryogenesis, slow-twitch muscle derives from the adaxial cells, a layer of paraxial mesoderm that differentiates medially within the myotome, immediately adjacent to the notochord. Subsequently, slow-twitch muscle cells migrate through the entire myotome, coming to lie at its most lateral surface. Here we examine the cellular and molecular basis for slow-twitch muscle cell migration. We show that slow-twitch muscle cell morphogenesis is marked by behaviors typical of cells influenced by differential cell adhesion. Dynamic and reciprocal waves of N-cadherin and M-cadherin expression within the myotome, which correlate precisely with cell migration, generate differential adhesive environments that drive slow-twitch muscle cell migration through the myotome. Removing or altering the expression of either protein within the myotome perturbs migration. These results provide a definitive example of homophilic cell adhesion shaping cellular behavior during vertebrate development.  相似文献   

19.
We describe a set of observations on developing zebrafish embryos and discuss the main conclusions they allow:(1) the embryonic dorso-ventral polarity axis is morphologically distinguishable prior to the onset of gastrulation; and (2) the involution of deep layer cells starts on the prospective dorsal side of the embryo. An asymmetry can be distinguished in the organization of the blastomeres in the zebrafish blastula at the 30% epiboly stage, in that one sector of the blastoderm is thicker than the other. Dye-labelling experiments with DiI and DiO and histological analysis allow us to conclude that the embryonic shield will form on the thinner side of the blastoderm. Therefore, this side corresponds to the prospective dorsal side of the embryo. Simultaneous injections of dyes on the thinner side of the blastoderm and on the opposite side show that involution of deep layer cells during gastrulation starts at the site at which the embryonic shield will form and extends from here to the prospective ventral regions of the germ ring.  相似文献   

20.
Directional cell migration is essential for almost all organisms during embryonic development, in adult life and contributes to pathological conditions. This is particularly critical during embryogenesis where it is essential that cells end up in their correct, precise locations in order to build a normal embryo. Many cells have solved this problem by following a gradient of a chemoattractant usually secreted by their target tissues. Our recent research has found an alternative, complimentary, mechanism where intracellular signals are able to generate cell polarity and directional migration in absence of any external chemoattactant. We used neural crest cells to study cell migration in vivo, by performing live imagining of the neural crest cell migrating during embryo development. We show that the Planar Cell Polarity (PCP) or non-canonical Wnt signaling pathway interacts with the proteoglycan syndecan-4 to control the direction in which cell protrusions are generated, and in consequence, the direction of migration. By analyzing the activity of the small GTPases using in vivo FRET imaging we showed that PCP signaling activates RhoA, while syndecan-4 inhibits Rac, both at the back of the neural crest cell. Here we discuss a model where these signals are integrated to generate directional migration in vivo.Key words: directional migration, cell migration, syndecan-4, PCP, non-canonical Wnt, neural crest, RhoA, RacThe ability of cells to move in a directed manner is a fundamental requirement for life. In multi-cellular organisms, this requirement begins in the embryo, where morphogenetic processes are dependent on the correct movement of large numbers of cells. In the adult too, cell migration plays a vital role in many systems including the immune system and wound healing. Cell migration defects can contribute to the pathology of many diseases including vascular diseases such as atherosclerosis, and chronic inflammatory diseases like asthma and multiple sclerosis. Likewise, metastasis in cancer is characterized by mis-regulation of the normal cell migration machinery and results in cells that are normally static becoming aggressively motile and invasive.Cell migration requires cell polarization and the formation of protrusions at one end of the cell. Polarization results in a different molecular ensemble at the front of the cell compared to that at the back. Cell protrusion formation at the front of the cell requires reorganization of the actin and microtubule cytoskeleton to produce a protrusion either in the form of a broad sheet-like lamellipodium or spiky filopodium. Small GTPases are well known modulators of these processes (reviewed in ref. 1).Several mechanism has been proposed as involved in directional migration during embryo development, such as chemotaxis (migration toward an soluble chemoattractant),2 haptotaxis (migration toward a substrate-bound chemoattractant),3 population pressure (migration from a region of high towards a region of low cell density)4 and contact inhibition of locomotion (change in the direction of migration as a consequence of cell-cell contact),5 being chemotaxis the most widely accepted and studied.The correct orientation of the cell and its protrusion is the keystone of directional migration and, in the case of chemotaxis, it is supposed to be controlled by the action of external chemical cues (chemoattractants) that are produced by or near to the target tissue.6 One of the best examples for chemoattraction in vivo is the migration of the progenitor germ cells, which are attracted by the chemokine SDF-1.2 It has been shown in vitro and in vivo, that upon receiving a chemotactic signal, the cell becomes polarized in the direction of migration. Nevertheless, it is known that cells cultured in vitro can became polarized and exhibit directional migration in absence of extrinsic chemoattractants.7 Pankov et al. showed that persistent directional migration in vitro can be achieved solely by modulating the activity of the small GTPase, Rac: high levels of Rac promotes the formation of peripheral lamella during random migration, while slightly lower levels of Rac suppress peripheral lamella and favour the formation of a polarized cell with lamella just at the leading edge.7 Is it possible that a similar mechanism of directional migration could occur in vivo?The migration of Neural Crest (NC) cells has been used as a model to study directional cell migration in vivo.810 The neural crest is an embryonic population of cells that are specified at the border between the neural plate and the epidermis.11 Upon induction neural crest cells undergo an epithelial to mesenchymal transition,12 detach from the neural tube and migrate following defined pathways that eventually allow them to colonize almost the entire embryo.13 Finally, after reaching their destination NC cells differentiate to form many different cell types including neurons, glia, cartilage, skeleton and pigment cells.14 The migration of the NC cells is critical for the proper differentiation of their derivatives and there are several human syndromes associated with failures in this process.The migration of NC cells is a highly ordered process; individual NC cells migrate with high persistence towards the direction of their targets,8 but until now it was not known how this directionality is controlled. A number of molecules have been identified as key players in neural crest migration, such as Ephrins, Semaphorins, Slit/Robo, etc. (reviewed in ref. 13). However most of these molecules work as inhibitory signals, which are required to restrict the migration of NC cells from prohibited areas. Although chemoattraction has been one of the proposed mechanisms to explain this directional migration, no chemoattractant has thus far been found in the NC.It has been known for many years that NC cells can migrate in vitro with a high directionality even in the absence of external signals.15 Therefore, our work has been focused on understanding how NC directionality is controlled. Recently, we have unveiled some of the molecules that control this directional migration in vitro. More importantly, we have been able to show that the same molecular machinery controls directional migration in vivo.9,10One of the key factors that controls directional migration of NC cells is the Planar Cell Polarity (PCP) or non-canonical Wnt signaling pathway.9,10,16 PCP signaling was first described in Drosophila, where a number of mutations were identified that disrupt the formation of bristles and hairs on the adult cuticle.17 In the Drosophila wing, epithelial cells are highly polarized, with a single hair outgrowth forming at the distal end of each cell. Mutations in PCP genes cause loss in cell polarity in this tissue with hairs forming in a disorganized pattern.18 In vertebrates, PCP signaling also regulates cell polarity during a number of different developmental processes including neural tube closure, cochlear hair orientation and ciliogenesis.19We have shown that the PCP pathway is essential for correct neural crest migration in Xenopus. Injection of dominant negative forms of the intracellular PCP component Dishevelled (Dsh), which inhibit the PCP pathway but not canonical Wnt signaling, block the migration of cranial neural crest cells in vivo.9 Recently this role has also been extended to zebrafish where directional migration of neural crest is severely disrupted in the PCP mutant trilobite (strabismus) and in embryos injected with a dominant negative form of Dsh or a morpholino against wnt5a,10 with no effect in neural crest cell motility.9,10 Two factors, pescadillo and syndecan-4 that have recently been proposed as modulators of the PCP signaling,20,21 are also required for NC migration.10,21 Taken together, these data point to an essential role for PCP signaling in neural crest migration.What is the cellular and molecular mechanism by which PCP signaling controls migration of NC cells? In order to investigate this question we analyzed the direction of neural crest migration and cell polarity in vitro and in vivo after interfering with two elements of the PCP signaling pathway: syndecan-4 and Dsh. One of the key finding of our work was that the inhibition of NC migration through syndecan-4 depletion does not affect the velocity of cell migration, but significantly reduces the directional migration of the cells in vivo (Fig. 1A and B). Consequently, when the orientation of cell protrusions was analyzed we found that syndecan-4 depletion does not affect the formation of cell protrusions, but the direction in which the cell protrusions are generated during migration. More precisely, normal cells extend their lamellipodia at the front of the cell (Fig. 1D), while cells where syndecan-4 is inhibited generate protrusion in all directions (Fig. 1E). A similar analysis was performed for embryos expressing a mutated form of Dsh that works as a dominant negative of PCP signaling and an equivalent effect on directional migration and the orientations of cell protrusions was observed (Fig. 1C and F).Open in a separate windowFigure 1Directional migration of neural crest cells. (A and B) Example of track of a single cell migrating in vivo. (A) Control cell showing persistent directional migration. (B) Cell in which the PCP signaling has been inhibited, showing absence of directional migration. (C) Cell in which syndecan-4 has been inhibited, showing no persistent migration. (D–F) Analysis of cell polarity and model of directional migration. Fn: fibronectin; Syn4: syndecan-4. (D) Control cell. Activation of Fn/Syn4 and PCP/RhoA lead to inhibition of Rac at the back of the cell, with the consequence polarization and directional migration. (E) Inhibition of PCP signaling leads to absence of RhoA activity, and in consequence an increase of Rac activity at the back of the cell. It seems that the inhibition of Rac activity by Syn4 is not sufficient to keep low levels of Rac at the back of the cells. High levels of Rac at the back produce a loss in cell polarity and in directional migration. (F) Inhibition of Syn4 generates high levels of Rac activity by a double mechanism: absence of direct inhibition of Rac and absence of RhoA which is dependent on PCP signaling. High levels of Rac at the back produce a loss of cell polarity and directional migration.As cell protrusions are known to be controlled by small GTPases and as PCP and syndecan-4 signaling regulates the activities of small GTPases,18,22 we analyzed the activity of cdc42, RhoA and Rac after interfering with Dsh and syndecan-4. We choose to perform FRET analysis of these molecules as it is a technique that allows the visualization of their localized activity. More interestingly we succeeded in performing FRET analysis in cells migrating in vivo for the first time. Our results show that syndecan-4 inhibits Rac activity, while Dsh signaling promotes RhoA activity. In addition, we show that RhoA inhibits Rac in neural crest cells.10 The regulation of Rac by syndecan-4 is similar to that seen in other cells types in vitro.23,24The model that emerges from these results to explain directional migration of NC cells in vivo is as follows (Fig. 1D). After delamination NC cells come into contact with fibronectin in the extracellular matrix, which is known to provide the main substrate for neural crest cells during their migration.25,26 The interaction of fibronectin with syndecan-4 leads to two major changes in the cell: activation of PCP signaling and inhibition of Rac activity. The activated PCP signaling becomes localized at the back of the cell. From here, PCP contributes to the inhibition of Rac at the back of the cell, through the activation of RhoA. The coordinated activities of syndecan-4 and PCP signaling lead to polarised Rac activity across the cell, with Rac enriched at the leading edge, where it promotes the polymerization of actin and formation of lamellipodia, resulting in directional migration (Fig. 1D). Inhibition of PCP signaling produces high levels of Rac all over the cell as Rac, an inhibitor of RhoA in many cell types including neural crest cells, is absent (Fig. 1E). This generates cell protrusions in all directions with the consequent loss of cell polarity. If syndecan-4 is absent, the levels of Rac activity are also high all over the cell as the inhibition of Rac by syndecan-4 is absent (Fig. 1F), which also leads to a loss of cell polarity.Although detailed study of the localized activity of small GTPases has not been performed for other migratory cells in vivo, it is likely that the machinery will be similar to the one described here for NC cells. For example, it is well established in Xenopus, zebrafish and chick embryos that the migration of mesodermal cells during gastrulation requires PCP signaling.2729 It has also been shown that gastrulation in Xenopus20 and in zebrafish (unpublished observations) requires the activity of syndecan-4. Thus, it is expected that cell polarity established during the migration of mesodermal cells will be dependent on small GTPases controlled by non-canonical Wnt signaling and syndecan-4.This novel integrated view of PCP, syndecan-4 and small GTPase activity during directional cell migration in vivo is an important advance in our knowledge of cell migration. Nevertheless, how the PCP signaling becomes activated only at the back of the cell, is a key question that needs to be answered. Future studies will be necessary to solve this and other crucial problems.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号