Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Elife ; 122023 04 19.
Article in English | MEDLINE | ID: mdl-37073859

ABSTRACT

Collective cell migration plays an essential role in vertebrate development, yet the extent to which dynamically changing microenvironments influence this phenomenon remains unclear. Observations of the distribution of the extracellular matrix (ECM) component fibronectin during the migration of loosely connected neural crest cells (NCCs) lead us to hypothesize that NCC remodeling of an initially punctate ECM creates a scaffold for trailing cells, enabling them to form robust and coherent stream patterns. We evaluate this idea in a theoretical setting by developing an individual-based computational model that incorporates reciprocal interactions between NCCs and their ECM. ECM remodeling, haptotaxis, contact guidance, and cell-cell repulsion are sufficient for cells to establish streams in silico, however, additional mechanisms, such as chemotaxis, are required to consistently guide cells along the correct target corridor. Further model investigations imply that contact guidance and differential cell-cell repulsion between leader and follower cells are key contributors to robust collective cell migration by preventing stream breakage. Global sensitivity analysis and simulated gain- and loss-of-function experiments suggest that long-distance migration without jamming is most likely to occur when leading cells specialize in creating ECM fibers, and trailing cells specialize in responding to environmental cues by upregulating mechanisms such as contact guidance.


Subject(s)
Fibronectins , Neural Crest , Cell Movement , Cell Communication
2.
Dev Dyn ; 252(5): 629-646, 2023 05.
Article in English | MEDLINE | ID: mdl-36692868

ABSTRACT

BACKGROUND: Collective and discrete neural crest cell (NCC) migratory streams are crucial to vertebrate head patterning. However, the factors that confine NCC trajectories and promote collective cell migration remain unclear. RESULTS: Computational simulations predicted that confinement is required only along the initial one-third of the cranial NCC migratory pathway. This guided our study of Colec12 (Collectin-12, a transmembrane scavenger receptor C-type lectin) and Trail (tumor necrosis factor-related apoptosis-inducing ligand, CD253) which we show expressed in chick cranial NCC-free zones. NCC trajectories are confined by Colec12 or Trail protein stripes in vitro and show significant and distinct changes in cell morphology and dynamic migratory characteristics when cocultured with either protein. Gain- or loss-of-function of either factor or in combination enhanced NCC confinement or diverted cell trajectories as observed in vivo with three-dimensional confocal microscopy, respectively, resulting in disrupted collective migration. CONCLUSIONS: These data provide evidence for Colec12 and Trail as novel NCC microenvironmental factors playing a role to confine cranial NCC trajectories and promote collective cell migration.


Subject(s)
Cell Movement , Chickens , Neural Crest , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Chickens/genetics , Chickens/physiology , Computer Simulation , Neural Crest/cytology , Neural Crest/physiology , Skull
3.
Development ; 148(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-35020873

ABSTRACT

The dynamics of multipotent neural crest cell differentiation and invasion as cells travel throughout the vertebrate embryo remain unclear. Here, we preserve spatial information to derive the transcriptional states of migrating neural crest cells and the cellular landscape of the first four chick cranial to cardiac branchial arches (BA1-4) using label-free, unsorted single-cell RNA sequencing. The faithful capture of branchial arch-specific genes led to identification of novel markers of migrating neural crest cells and 266 invasion genes common to all BA1-4 streams. Perturbation analysis of a small subset of invasion genes and time-lapse imaging identified their functional role to regulate neural crest cell behaviors. Comparison of the neural crest invasion signature to other cell invasion phenomena revealed a shared set of 45 genes, a subset of which showed direct relevance to human neuroblastoma cell lines analyzed after exposure to the in vivo chick embryonic neural crest microenvironment. Our data define an important spatio-temporal reference resource to address patterning of the vertebrate head and neck, and previously unidentified cell invasion genes with the potential for broad impact.


Subject(s)
Branchial Region/growth & development , Head/growth & development , Neck/growth & development , Neural Crest/growth & development , Animals , Body Patterning/genetics , Branchial Region/embryology , Cell Differentiation/genetics , Cell Movement/genetics , Cellular Microenvironment/genetics , Chick Embryo , Embryo, Mammalian , Embryo, Nonmammalian , Embryonic Development/genetics , Head/embryology , Humans , Mesoderm/growth & development , Multipotent Stem Cells/cytology , Neck/embryology , Neural Crest/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Organogenesis/genetics , Tumor Microenvironment/genetics , Vertebrates/genetics , Vertebrates/growth & development
4.
Development ; 147(1)2020 01 09.
Article in English | MEDLINE | ID: mdl-31826865

ABSTRACT

Neural crest migration requires cells to move through an environment filled with dense extracellular matrix and mesoderm to reach targets throughout the vertebrate embryo. Here, we use high-resolution microscopy, computational modeling, and in vitro and in vivo cell invasion assays to investigate the function of Aquaporin 1 (AQP-1) signaling. We find that migrating lead cranial neural crest cells express AQP-1 mRNA and protein, implicating a biological role for water channel protein function during invasion. Differential AQP-1 levels affect neural crest cell speed and direction, as well as the length and stability of cell filopodia. Furthermore, AQP-1 enhances matrix metalloprotease activity and colocalizes with phosphorylated focal adhesion kinases. Colocalization of AQP-1 with EphB guidance receptors in the same migrating neural crest cells has novel implications for the concept of guided bulldozing by lead cells during migration.


Subject(s)
Aquaporin 1/physiology , Cell Movement/physiology , Neural Crest/cytology , Pseudopodia/physiology , Animals , Branchial Region/cytology , Branchial Region/embryology , Cell Membrane/physiology , Cellular Microenvironment , Chick Embryo , Computational Biology , Focal Adhesions , Neural Crest/embryology , Receptor, EphB1/metabolism , Receptor, EphB3/metabolism
5.
Elife ; 62017 12 04.
Article in English | MEDLINE | ID: mdl-29199959

ABSTRACT

Neural crest cells migrate throughout the embryo, but how cells move in a directed and collective manner has remained unclear. Here, we perform the first single-cell transcriptome analysis of cranial neural crest cell migration at three progressive stages in chick and identify and establish hierarchical relationships between cell position and time-specific transcriptional signatures. We determine a novel transcriptional signature of the most invasive neural crest Trailblazer cells that is consistent during migration and enriched for approximately 900 genes. Knockdown of several Trailblazer genes shows significant but modest changes to total distance migrated. However, in vivo expression analysis by RNAscope and immunohistochemistry reveals some salt and pepper patterns that include strong individual Trailblazer gene expression in cells within other subregions of the migratory stream. These data provide new insights into the molecular diversity and dynamics within a neural crest cell migratory stream that underlie complex directed and collective cell behaviors.


Subject(s)
Cell Movement , Gene Expression Profiling , Neural Crest/physiology , Single-Cell Analysis , Animals , Chick Embryo , Spatio-Temporal Analysis
6.
J Cell Biol ; 216(10): 3339-3354, 2017 10 02.
Article in English | MEDLINE | ID: mdl-28811280

ABSTRACT

Neural crest cells are both highly migratory and significant to vertebrate organogenesis. However, the signals that regulate neural crest cell migration remain unclear. In this study, we test the function of differential screening-selected gene aberrant in neuroblastoma (DAN), a bone morphogenetic protein (BMP) antagonist we detected by analysis of the chick cranial mesoderm. Our analysis shows that, before neural crest cell exit from the hindbrain, DAN is expressed in the mesoderm, and then it becomes absent along cell migratory pathways. Cranial neural crest and metastatic melanoma cells avoid DAN protein stripes in vitro. Addition of DAN reduces the speed of migrating cells in vivo and in vitro, respectively. In vivo loss of function of DAN results in enhanced neural crest cell migration by increasing speed and directionality. Computer model simulations support the hypothesis that DAN restrains cell migration by regulating cell speed. Collectively, our results identify DAN as a novel factor that inhibits uncontrolled neural crest and metastatic melanoma invasion and promotes collective migration in a manner consistent with the inhibition of BMP signaling.


Subject(s)
Avian Proteins/metabolism , Cell Movement , Chickens/metabolism , Melanoma/metabolism , Neural Crest/embryology , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Avian Proteins/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Chick Embryo , Chickens/genetics , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Neural Crest/pathology , Tumor Suppressor Proteins/genetics
7.
Dev Biol ; 407(1): 12-25, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26278036

ABSTRACT

Embryonic neural crest cells travel in discrete streams to precise locations throughout the head and body. We previously showed that cranial neural crest cells respond chemotactically to vascular endothelial growth factor (VEGF) and that cells within the migratory front have distinct behaviors and gene expression. We proposed a cell-induced gradient model in which lead neural crest cells read out directional information from a chemoattractant profile and instruct trailers to follow. In this study, we show that migrating chick neural crest cells do not display distinct lead and trailer gene expression profiles in culture. However, exposure to VEGF in vitro results in the upregulation of a small subset of genes associated with an in vivo lead cell signature. Timed addition and removal of VEGF in culture reveals the changes in neural crest cell gene expression are rapid. A computational model incorporating an integrate-and-switch mechanism between cellular phenotypes predicts migration efficiency is influenced by the timescale of cell behavior switching. To test the model hypothesis that neural crest cellular phenotypes respond to changes in the VEGF chemoattractant profile, we presented ectopic sources of VEGF to the trailer neural crest cell subpopulation and show diverted cell trajectories and stream alterations consistent with model predictions. Gene profiling of trailer cells that diverted and encountered VEGF revealed upregulation of a subset of 'lead' genes. Injection of neuropilin1 (Np1)-Fc into the trailer subpopulation or electroporation of VEGF morpholino to reduce VEGF signaling failed to alter trailer neural crest cell trajectories, suggesting trailers do not require VEGF to maintain coordinated migration. These results indicate that VEGF is one of the signals that establishes lead cell identity and its chemoattractant profile is critical to neural crest cell migration.


Subject(s)
Neural Crest/cytology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Movement , Cellular Microenvironment , Chemotaxis , Chick Embryo , Computer Simulation , Gene Expression Regulation, Developmental
8.
Development ; 142(11): 2014-25, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25977364

ABSTRACT

Neural crest (NC) cell migration is crucial to the formation of peripheral tissues during vertebrate development. However, how NC cells respond to different microenvironments to maintain persistence of direction and cohesion in multicellular streams remains unclear. To address this, we profiled eight subregions of a typical cranial NC cell migratory stream. Hierarchical clustering showed significant differences in the expression profiles of the lead three subregions compared with newly emerged cells. Multiplexed imaging of mRNA expression using fluorescent hybridization chain reaction (HCR) quantitatively confirmed the expression profiles of lead cells. Computational modeling predicted that a small fraction of lead cells that detect directional information is optimal for successful stream migration. Single-cell profiling then revealed a unique molecular signature that is consistent and stable over time in a subset of lead cells within the most advanced portion of the migratory front, which we term trailblazers. Model simulations that forced a lead cell behavior in the trailing subpopulation predicted cell bunching near the migratory domain entrance. Misexpression of the trailblazer molecular signature by perturbation of two upstream transcription factors agreed with the in silico prediction and showed alterations to NC cell migration distance and stream shape. These data are the first to characterize the molecular diversity within an NC cell migratory stream and offer insights into how molecular patterns are transduced into cell behaviors.


Subject(s)
Cell Movement , Gene Expression Profiling , Gene Expression Regulation, Developmental , Neural Crest/cytology , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Movement/genetics , Chick Embryo , Computer Simulation , Gene Knockdown Techniques , Neural Crest/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Single-Cell Analysis
9.
Development ; 141(5): 1095-103, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550117

ABSTRACT

Embryonic cells that migrate long distances must critically balance cell division in order to maintain stream dynamics and population of peripheral targets. Yet details of individual cell division events and how cell cycle is related to phases of migration remain unclear. Here, we examined these questions using the chick cranial neural crest (NC). In vivo time-lapse imaging revealed that a typical migrating NC cell division event lasted ~1 hour and included four stereotypical steps. Cell tracking showed that dividing NC cells maintained position relative to non-dividing neighbors. NC cell division orientation and the time and distance to first division after neural tube exit were stochastic. To address how cell cycle is related to phases of migration, we used FACs analysis to identify significant spatiotemporal differences in NC cell cycle profiles. Two-photon photoconversion of single and small numbers of mKikGR-labeled NC cells confirmed that lead NC cells exhibited a nearly fourfold faster doubling time after populating the branchial arches. By contrast, Ki-67 staining showed that one out of every five later emerging NC cells exited the cell cycle after reaching proximal head targets. The relatively quiescent mitotic activity during NC cell migration to the branchial arches was altered when premigratory cells were reduced in number by tissue ablation. Together, our results provide the first comprehensive details of the pattern and dynamics of cell division events during cranial NC cell migration.


Subject(s)
Neural Crest/cytology , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Division/genetics , Cell Division/physiology , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Chick Embryo , Flow Cytometry , Neural Crest/metabolism
10.
BMC Dev Biol ; 10: 101, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20868502

ABSTRACT

BACKGROUND: Tracing cell dynamics in the embryo becomes tremendously difficult when cell trajectories cross in space and time and tissue density obscure individual cell borders. Here, we used the chick neural crest (NC) as a model to test multicolor cell labeling and multispectral confocal imaging strategies to overcome these roadblocks. RESULTS: We found that multicolor nuclear cell labeling and multispectral imaging led to improved resolution of in vivo NC cell identification by providing a unique spectral identity for each cell. NC cell spectral identity allowed for more accurate cell tracking and was consistent during short term time-lapse imaging sessions. Computer model simulations predicted significantly better object counting for increasing cell densities in 3-color compared to 1-color nuclear cell labeling. To better resolve cell contacts, we show that a combination of 2-color membrane and 1-color nuclear cell labeling dramatically improved the semi-automated analysis of NC cell interactions, yet preserved the ability to track cell movements. We also found channel versus lambda scanning of multicolor labeled embryos significantly reduced the time and effort of image acquisition and analysis of large 3D volume data sets. CONCLUSIONS: Our results reveal that multicolor cell labeling and multispectral imaging provide a cellular fingerprint that may uniquely determine a cell's position within the embryo. Together, these methods offer a spectral toolbox to resolve in vivo cell dynamics in unprecedented detail.


Subject(s)
Cell Lineage , Chick Embryo/cytology , Computer Simulation , Image Interpretation, Computer-Assisted , Neural Crest/cytology , Animals , Microscopy, Confocal
11.
Dev Biol ; 339(1): 114-25, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20036652

ABSTRACT

The neural crest is an excellent model to study embryonic cell migration, since cell behaviors can be studied in vivo with advanced optical imaging and molecular intervention. What is unclear is how molecular signals direct neural crest cell (NCC) migration through multiple microenvironments and into specific targets. Here, we tested the hypothesis that the invasion of cranial NCCs, specifically the rhombomere 4 (r4) migratory stream into branchial arch 2 (ba2), is due to chemoattraction through neuropilin-1-vascular endothelial growth factor (VEGF) interactions. We found that the spatio-temporal expression pattern of VEGF in the ectoderm correlated with the NCC migratory front. RT-PCR analysis of the r4 migratory stream showed that ba2 tissue expressed VEGF and r4 NCCs expressed VEGF receptor 2. When soluble VEGF receptor 1 (sVEGFR1) was injected distal to the r4 migratory front, to bind up endogenous VEGF, NCCs failed to completely invade ba2. Time-lapse imaging revealed that cranial NCCs were attracted to ba2 tissue or VEGF sources in vitro. VEGF-soaked beads or VEGF-expressing cells placed adjacent to the r4 migratory stream caused NCCs to divert from stereotypical pathways and move towards an ectopic VEGF source. Our results suggest a model in which NCC entry and invasion of ba2 is dependent on chemoattractive signaling through neuropilin-1-VEGF interactions.


Subject(s)
Cell Movement , Neural Crest/cytology , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Proliferation , Chick Embryo , Immunohistochemistry , In Situ Hybridization , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
12.
Dev Dyn ; 237(10): 2657-66, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18629870

ABSTRACT

The embryonic microenvironment is an important source of signals that program multipotent cells to adopt a particular fate and migratory path, yet its potential to reprogram and restrict multipotent tumor cell fate and invasion is unrealized. Aggressive tumor cells share many characteristics with multipotent, invasive embryonic progenitors, contributing to the paradigm of tumor cell plasticity. In the vertebrate embryo, multiple cell types originate from a highly invasive cell population called the neural crest. The neural crest and the embryonic microenvironments they migrate through represent an excellent model system to study cell diversification during embryogenesis and phenotype determination. Recent exciting studies of tumor cells transplanted into various embryo models, including the neural crest rich chick microenvironment, have revealed the potential to control and revert the metastatic phenotype, suggesting further work may help to identify new targets for therapeutic intervention derived from a convergence of tumorigenic and embryonic signals. In this mini-review, we summarize markers that are common to the neural crest and highly aggressive human melanoma cells. We highlight advances in our understanding of tumor cell behaviors and plasticity studied within the chick neural crest rich microenvironment. In so doing, we honor the tremendous contributions of Professor Elizabeth D. Hay toward this important interface of developmental and cancer biology.


Subject(s)
Neoplastic Stem Cells/cytology , Neural Crest/cytology , Neural Crest/embryology , Animals , Cell Lineage , Cell Movement , Humans , Melanoma/pathology , Models, Biological
13.
Dev Biol ; 316(2): 275-87, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18328476

ABSTRACT

Neural crest cell (NCC) invasion is a complex sculpting of individual cells into organized migratory streams that lead to organ development along the vertebrate axis. Key to our understanding of how molecular mechanisms modulate the NCC migratory pattern is information about cell behaviors, yet it has been challenging to selectively mark and analyze migratory NCCs in a living embryo. Here, we apply an innovative in vivo strategy to investigate chick NCC behaviors within the rhombomere 4 (r4) migratory stream by combining photoactivation of KikGR and confocal time-lapse analysis of H2B-mRFP1 transfected NCCs. We find that the spatial order of r4 NCC emergence translates into a distal-to-proximal invasion of the 2nd branchial arch. Lead and trailing NCCs display similar average cell speeds and directionalities. Surprisingly, we find that lead NCCs proliferate along the migratory route and grow to outnumber trailing NCCs by nearly 3 to 1. A simple, cell-based computational model reproduces the r4 NCC migratory pattern and predicts the invasion order can be disrupted by slower, less directional lead cells or by environmental noise. Our results suggest a model in which NCC behaviors maintain a spatially-ordered invasion of the branchial arches with differences in cell proliferation between the migratory front and trailing NCCs.


Subject(s)
Brain/embryology , Chick Embryo/physiology , Luminescent Proteins/genetics , Neural Crest/cytology , Neural Crest/physiology , Animals , Cell Division , Cell Movement , Head/embryology , Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 103(10): 3752-7, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16505384

ABSTRACT

Human metastatic melanoma cells express a dedifferentiated, plastic phenotype, which may serve as a selective advantage, because melanoma cells invade various microenvironments. Over the last three decades, there has been an increased focus on the role of the tumor microenvironment in cancer progression, with the goal of reversing the metastatic phenotype. Here, using an embryonic chick model, we explore the possibility of reverting the metastatic melanoma phenotype to its cell type of origin, the neural-crest-derived melanocyte. GFP-labeled adult human metastatic melanoma cells were transplanted in ovo adjacent to host chick premigratory neural crest cells and analyzed 48 and 96 h after egg reincubation. Interestingly, the transplanted melanoma cells do not form tumors. Instead, we find that transplanted melanoma cells invade surrounding chick tissues in a programmed manner, distributing along host neural-crest-cell migratory pathways. The invading melanoma cells display neural-crest-cell-like morphologies and populate host peripheral structures, including the branchial arches, dorsal root and sympathetic ganglia. Analysis of a melanocyte-specific phenotype marker (MART-1) and a neuronal marker (Tuj1) revealed a subpopulation of melanoma cells that invade the chick periphery and express MART-1 and Tuj1. Our results demonstrate the ability of adult human metastatic melanoma cells to respond to chick embryonic environmental cues, a subset of which may undergo a reprogramming of their metastatic phenotype. This model has the potential to provide insights into the regulation of tumor cell plasticity by an embryonic milieu, which may hold significant therapeutic promise.


Subject(s)
Melanoma, Experimental/secondary , Animals , Antigens, Neoplasm , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Line, Tumor , Chick Embryo , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Humans , MART-1 Antigen , Melanocytes/cytology , Melanocytes/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neural Crest/cytology , Neural Crest/embryology , Phenotype , Tubulin/metabolism
15.
Biotechniques ; 39(5): 703-10, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16312219

ABSTRACT

Our understanding of the molecular mechanisms that direct cell motility, cell division, and cell shaping has benefited from innovations in cell labeling and the ability to resolve intracellular dynamics with multispectral, high-resolution imaging. However, due to difficulties with in vivo cell marking and monitoring, most studies have been restricted to fixed tissue or cells in culture. Here, we report the delivery of multiple (up to four), multicolor fluorescent protein (FP) constructs and four-dimensional (4-D), multispectral time-lapse confocal imaging of cell movements in living chick embryos. Cell cytoskeletal components are fluorescently tagged after microinjection and electroporation of a cocktail of FP constructs into specific regions of chick embryos. We tested 11 different FP constructs in various two-, three-, and four-color combinations using multispectral imaging and linear unmixing to limit the crosstalk between different emission spectra. We monitored intracellular dynamics in individual multicolored migrating cells in vivo and developed a set of advantageous imaging parameters for 4-D time-lapse confocal microscopy. We find that the number of four-color labeled cells in a typical embryo is approximately 10% of the total number of fluorescently labeled cells; this value consistently increases showing that approximately 50% of the total labeled cells have only one-color. We find that multicolored cells are photostable for time-lapses of approximately 2-3 h. Thus, cell labeling with up to four FP color schemes combined with multispectral, 4-D confocal time-lapse imaging offers a powerful tool to simultaneously analyze cellular and molecular dynamics during chick embryogenesis.


Subject(s)
Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Animals , Cell Division , Cell Movement , Cell Shape , Chick Embryo , Color , Cytoskeleton/metabolism , Electroporation , Fluorescent Dyes/pharmacology , Genetic Techniques , Genetic Vectors , Green Fluorescent Proteins/metabolism , Light , Luminescent Proteins/metabolism , Photobleaching , Photons , Recombinant Fusion Proteins/chemistry , Time Factors
16.
Development ; 131(24): 6141-51, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15548586

ABSTRACT

The proper assembly of craniofacial structures and the peripheral nervous system requires neural crest cells to emerge from the neural tube and navigate over long distances to the branchial arches. Cell and molecular studies have shed light on potential intrinsic and extrinsic cues, which, in combination, are thought to ensure the induction and specification of cranial neural crest cells. However, much less is known about how migrating neural crest cells interpret and integrate signals from the microenvironment and other neural crest cells to sort into and maintain the stereotypical pattern of three spatially segregated streams. Here, we explore the extent to which cranial neural crest cells use cell-to-cell and cell-environment interactions to pathfind. The cell membrane and cytoskeletal elements in chick premigratory neural crest cells were labeled in vivo. Three-dimensional reconstructions of migrating neural crest cells were then obtained using confocal static and time-lapse imaging. It was found that neural crest cells maintained nearly constant contact with other migrating neural crest cells, in addition to the microenvironment. Cells used lamellipodia or short, thin filopodia (1-2 microm wide) for local contacts (<20 microm). Non-local, long distance contact (up to 100 microm) was initiated by filopodia that extended and retracted, extended and tracked, or tethered two non-neighboring cells. Intriguingly, the cell-to-cell contacts often stimulated a cell to change direction in favor of a neighboring cell's trajectory. In summary, our results present in vivo evidence for local and long-range neural crest cell interactions, suggesting a possible role for these contacts in directional guidance.


Subject(s)
Cell Communication/physiology , Cytoskeleton/metabolism , Neural Crest/cytology , Neurons/cytology , Skull/cytology , Animals , Chick Embryo , Microscopy, Confocal , Neural Crest/growth & development , Pseudopodia/metabolism , Skull/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...