Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
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(8): 1130-1142, 2023 08.
Article in English | MEDLINE | ID: mdl-36840366

ABSTRACT

BACKGROUND: The molecular identification of neural progenitor cell populations that connect to establish the sympathetic nervous system (SNS) remains unclear. This is due to technical limitations in the acquisition and spatial mapping of molecular information to tissue architecture. RESULTS: To address this, we applied Slide-seq spatial transcriptomics to intact fresh frozen chick trunk tissue transversely cryo-sectioned at the developmental stage prior to SNS formation. In parallel, we performed age- and location-matched single cell (sc) RNA-seq and 10× Genomics Visium to inform our analysis. Downstream bioinformatic analyses led to the unique molecular identification of neural progenitor cells within the peripheral sympathetic ganglia (SG) and spinal cord preganglionic neurons (PGNs). We then successfully applied the HiPlex RNAscope fluorescence in situ hybridization and multispectral confocal microscopy to visualize 12 gene targets in stage-, age- and location-matched chick trunk tissue sections. CONCLUSIONS: Together, these data demonstrate a robust strategy to acquire and integrate single cell and spatial transcriptomic information, resulting in improved resolution of molecular heterogeneities in complex neural tissue architectures. Successful application of this strategy to the developing SNS provides a roadmap for functional studies of neural connectivity and platform to address complex questions in neural development and regeneration.


Subject(s)
Sympathetic Nervous System , Transcriptome , Animals , RNA, Messenger , In Situ Hybridization, Fluorescence , Ganglia, Sympathetic , Chickens
3.
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
4.
Dev Biol ; 480: 78-90, 2021 12.
Article in English | MEDLINE | ID: mdl-34416224

ABSTRACT

Mistakes in trunk neural crest (NC) cell migration may lead to birth defects of the sympathetic nervous system (SNS) and neuroblastoma (NB) cancer. Receptor tyrosine kinase B (TrkB) and its ligand BDNF critically regulate NC cell migration during normal SNS development and elevated expression of TrkB is correlated with high-risk NB patients. However, in the absence of a model with in vivo interrogation of human NB cell and gene expression dynamics, the mechanistic role of TrkB in NB disease progression remains unclear. Here, we study the functional relationship between TrkB, cell invasion and plasticity of human NB cells by taking advantage of our validated in vivo chick embryo transplant model. We find that LAN5 (high TrkB) and SHSY5Y (moderate TrkB) human NB cells aggressively invade host embryos and populate typical NC targets, however loss of TrkB function significantly reduces cell invasion. In contrast, NB1643 (low TrkB) cells remain near the transplant site, but over-expression of TrkB leads to significant cell invasion. Invasive NB cells show enhanced expression of genes indicative of the most invasive host NC cells. In contrast, transplanted human NB cells down-regulate known NB tumor initiating and stem cell markers. Human NB cells that remain within the dorsal neural tube transplant also show enhanced expression of cell differentiation genes, resulting in an improved disease outcome as predicted by a computational algorithm. These in vivo data support TrkB as an important biomarker and target to control NB aggressiveness and identify the chick embryonic trunk neural crest microenvironment as a source of signals to drive NB to a less aggressive state, likely acting at the dorsal neural tube.


Subject(s)
Membrane Glycoproteins/metabolism , Neoplasm Invasiveness/genetics , Neural Crest/embryology , Receptor, trkB/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Plasticity/genetics , Cell Transformation, Neoplastic/metabolism , Chick Embryo , Gene Expression , Humans , Membrane Glycoproteins/genetics , Neural Crest/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, trkB/genetics , Signal Transduction/genetics , Tumor Microenvironment/genetics
5.
Bull Math Biol ; 83(4): 26, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33594536

ABSTRACT

Cell invasion and cell plasticity are critical to human development but are also striking features of cancer metastasis. By distributing a multipotent cell type from a place of birth to distal locations, the vertebrate embryo builds organs. In comparison, metastatic tumor cells often acquire a de-differentiated phenotype and migrate away from a primary site to inhabit new microenvironments, disrupting normal organ function. Countless observations of both embryonic cell migration and tumor metastasis have demonstrated complex cell signaling and interactive behaviors that have long confounded scientist and clinician alike. James D. Murray realized the important role of mathematics in biology and developed a unique strategy to address complex biological questions such as these. His work offers a practical template for constructing clear, logical, direct and verifiable models that help to explain complex cell behaviors and direct new experiments. His pioneering work at the interface of development and cancer made significant contributions to glioblastoma cancer and embryonic pattern formation using often simple models with tremendous predictive potential. Here, we provide a brief overview of advances in cell invasion and cell plasticity using the embryonic neural crest and its ancestral relationship to aggressive cancers that put into current context the timeless aspects of his work.


Subject(s)
Models, Biological , Neoplasm Invasiveness , Neoplasms , Humans , Neoplasms/physiopathology , Neural Crest/cytology
6.
Methods Mol Biol ; 2179: 107-114, 2021.
Article in English | MEDLINE | ID: mdl-32939716

ABSTRACT

Live embryo imaging may provide a wealth of information on intact cell and tissue dynamics, but can be technically challenging to sustain embryo orientation and health for long periods under a microscope. In this protocol, we describe an in vivo method to mount and image cell movements during the epithelial-to-mesenchymal transition (EMT) of neural crest cells within the chick dorsal neural tube. We focus on describing the collection of images and data preparation for image analysis throughout the developmental stages HH15-21 in the chick trunk. Trunk neural crest cell EMT is crucial to development of the peripheral nervous system and pigment cell patterning. The methods we describe may also be applied to other cell and tissue phenomena at various chick developmental stages with some modifications.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Molecular Imaging/methods , Neural Crest/ultrastructure , Neural Tube/ultrastructure , Animals , Cell Movement/genetics , Chick Embryo , Neural Tube/growth & development
7.
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
8.
Dev Biol ; 461(2): 184-196, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32084354

ABSTRACT

Vertebrate head morphogenesis involves carefully-orchestrated tissue growth and cell movements of the mesoderm and neural crest to form the distinct craniofacial pattern. To better understand structural birth defects, it is important that we characterize the dynamics of these processes and learn how they rely on each other. Here we examine this question during chick head morphogenesis using time-lapse imaging, computational modeling, and experiments. We find that head mesodermal cells in culture move in random directions as individuals and move faster in the presence of neural crest cells. In vivo, mesodermal cells migrate in a directed manner and maintain neighbor relationships; neural crest cells travel through the mesoderm at a faster speed. The mesoderm grows with a non-uniform spatio-temporal profile determined by BrdU labeling during the period of faster and more-directed neural crest collective migration through this domain. We use computer simulations to probe the robustness of neural crest stream formation by varying the spatio-temporal growth profile of the mesoderm. We follow this with experimental manipulations that either stop mesoderm growth or prevent neural crest migration and observe changes in the non-manipulated cell population, implying a dynamic feedback between tissue growth and neural crest cell signaling to confer robustness to the system. Overall, we present a novel descriptive analysis of mesoderm and neural crest cell dynamics that reveals the coordination and co-dependence of these two cell populations during head morphogenesis.


Subject(s)
Chick Embryo/cytology , Head/embryology , Mesoderm/cytology , Neural Crest/cytology , Neural Tube/cytology , Animals , Cell Division , Cell Movement , Cells, Cultured , Chickens , Computer Simulation , Coturnix/embryology , Ectoderm/cytology , Models, Biological , Morphogenesis , Time-Lapse Imaging
9.
Dev Dyn ; 249(3): 270-280, 2020 03.
Article in English | MEDLINE | ID: mdl-31622517

ABSTRACT

The neural crest serves as a powerful and tractable model paradigm for understanding collective cell migration. The neural crest cell populations are well-known for their long-distance collective migration and contribution to diverse cell lineages during vertebrate development. If neural crest cells fail to reach a target or populate an incorrect location, then improper cell differentiation or uncontrolled cell proliferation can result. A wide range of interdisciplinary studies has been carried out to understand the response of neural crest cells to different stimuli and their ability to migrate to distant targets. In this critical commentary, we illustrate how an interdisciplinary collaboration involving experimental and mathematical modeling has led to a deeper understanding of cranial neural crest cell migration. We identify open questions and propose possible ways to start answering some of the challenges arising.


Subject(s)
Cell Movement/physiology , Neural Crest/cytology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Humans , Interdisciplinary Studies , Models, Theoretical , Neural Crest/metabolism , Signal Transduction/physiology
10.
J Math Biol ; 80(1-2): 481-504, 2020 01.
Article in English | MEDLINE | ID: mdl-31587096

ABSTRACT

A huge variety of mathematical models have been used to investigate collective cell migration. The aim of this brief review is twofold: to present a number of modelling approaches that incorporate the key factors affecting cell migration, including cell-cell and cell-tissue interactions, as well as domain growth, and to showcase their application to model the migration of neural crest cells. We discuss the complementary strengths of microscale and macroscale models, and identify why it can be important to understand how these modelling approaches are related. We consider neural crest cell migration as a model paradigm to illustrate how the application of different mathematical modelling techniques, combined with experimental results, can provide new biological insights. We conclude by highlighting a number of future challenges for the mathematical modelling of neural crest cell migration.


Subject(s)
Cell Movement/physiology , Models, Biological , Neural Crest/growth & development , Animals , Cell Communication/physiology , Cell Line, Tumor , Humans , Neural Crest/cytology , Xenopus , Zebrafish
11.
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
12.
Methods Mol Biol ; 1920: C1, 2019.
Article in English | MEDLINE | ID: mdl-31290130

ABSTRACT

The author added a sentence to this chapter. The text has been added to the chapter opening page.

13.
Methods Mol Biol ; 1976: 71-82, 2019.
Article in English | MEDLINE | ID: mdl-30977066

ABSTRACT

In ovo electroporation enables transfection of non-viral plasmid DNA and/or morpholinos to fluorescently label and/or perturb gene function in cells of interest. However, targeted electroporation into specific subregions of the embryo can be challenging due to placement and size limitations of the electrodes. Here we describe the basic techniques for in ovo electroporation in the chick embryo and suggest parameters to electroporate cells within different target tissues that with some modifications may be applicable to a wide range of developmental stages and other embryo model organisms.


Subject(s)
Electroporation/methods , Morpholinos/metabolism , Plasmids/genetics , Animals , Chick Embryo , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental
14.
Methods Mol Biol ; 1920: 247-263, 2019.
Article in English | MEDLINE | ID: mdl-30737695

ABSTRACT

Reptiles have great taxonomic diversity that is reflected in their morphology, ecology, physiology, modes of reproduction, and development. Interest in comparative and evolutionary developmental biology makes protocols for the study of reptile embryos invaluable resources. The relatively large size, seasonal breeding, and long gestation times of turtles epitomize the challenges faced by the developmental biologist. We describe protocols for the preparation of turtle embryos for ex ovo culture, electroporation, in situ hybridization, and microcomputed tomography. Because these protocols have been adapted and optimized from methods used for frog, chick, and mouse embryos, it is likely that they could be used for other reptilian species. Notes are included for alligator embryos where appropriate.


Subject(s)
Alligators and Crocodiles/embryology , Embryonic Development , Turtles/embryology , Alligators and Crocodiles/genetics , Animals , Biomarkers , Electroporation , Embryo Culture Techniques , Embryonic Development/genetics , Gene Expression Regulation, Developmental , In Situ Hybridization , Turtles/genetics , X-Ray Microtomography
15.
Genesis ; 56(9): e23239, 2018 09.
Article in English | MEDLINE | ID: mdl-30133140

ABSTRACT

Trunk neural crest cells follow a common ventral migratory pathway but are distributed into two distinct locations to form discrete sympathetic and dorsal root ganglia along the vertebrate axis. Although fluorescent cell labeling and time-lapse studies have recorded complex trunk neural crest cell migratory behaviors, the signals that underlie this dynamic patterning remain unclear. The absence of molecular information has led to a number of mechanistic hypotheses for trunk neural crest cell migration. Here, we review recent data in support of three distinct mechanisms of trunk neural crest cell migration and develop and simulate a computational model based on chemotactic signaling. We show that by integrating the timing and spatial location of multiple chemotactic signals, trunk neural crest cells may be accurately positioned into two distinct targets that correspond to the sympathetic and dorsal root ganglia. In doing so, we honor the contributions of Wilhelm His to his identification of the neural crest and extend the observations of His and others to better understand a complex question in neural crest cell biology.


Subject(s)
Cell Movement , Chemotaxis , Models, Biological , Neural Crest/cytology , Animals , Chemokines/physiology , Endothelial Cells/cytology , Humans , Signal Transduction
16.
Biophys Chem ; 238: 30-38, 2018 07.
Article in English | MEDLINE | ID: mdl-29734136

ABSTRACT

Genomic information from human patient samples of pediatric neuroblastoma cancers and known outcomes have led to specific gene lists put forward as high risk for disease progression. However, the reliance on gene expression correlations rather than mechanistic insight has shown limited potential and suggests a critical need for molecular network models that better predict neuroblastoma progression. In this study, we construct and simulate a molecular network of developmental genes and downstream signals in a 6-gene input logic model that predicts a favorable/unfavorable outcome based on the outcome of the four cell states including cell differentiation, proliferation, apoptosis, and angiogenesis. We simulate the mis-expression of the tyrosine receptor kinases, trkA and trkB, two prognostic indicators of neuroblastoma, and find differences in the number and probability distribution of steady state outcomes. We validate the mechanistic model assumptions using RNAseq of the SHSY5Y human neuroblastoma cell line to define the input states and confirm the predicted outcome with antibody staining. Lastly, we apply input gene signatures from 77 published human patient samples and show that our model makes more accurate disease outcome predictions for early stage disease than any current neuroblastoma gene list. These findings highlight the predictive strength of a logic-based model based on developmental genes and offer a better understanding of the molecular network interactions during neuroblastoma disease progression.


Subject(s)
Logic , Models, Biological , Neuroblastoma/genetics , Cell Line, Tumor , Humans , Neuroblastoma/metabolism
17.
Dev Biol ; 444 Suppl 1: S352-S355, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29660313

ABSTRACT

The embryonic microenvironment is an important source of signals that promote multipotent cells to adopt a specific fate and direct cells along distinct migratory pathways. Yet, the ability of the embryonic microenvironment to retain multipotent progenitors or reprogram de-differentiated cells is less clear. Mistakes in cell differentiation or migration often result in developmental defects and tumorigenesis, including aggressive cancers that share many characteristics with embryonic progenitor cells. This is a striking feature of the vertebrate neural crest, a multipotent and highly migratory cell population first identified by His (1868) with the potential to metamorphose into aggressive melanoma cancer. In this perspective, we address the roles of CD271/p75 in tumor initiation, phenotype switching and reprogramming of metastatic melanoma and discuss the convergence of these roles in melanoma plasticity.


Subject(s)
Adapalene/metabolism , Adaptor Proteins, Signal Transducing/physiology , Melanoma/metabolism , Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic/metabolism , Embryonic Stem Cells/physiology , Humans , Melanocytes/cytology , Melanoma/physiopathology , Mice , Multipotent Stem Cells , Neural Crest/embryology , Neural Crest/metabolism , Neural Crest/physiology , Neuronal Plasticity/physiology
18.
Biol Open ; 7(1)2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29175861

ABSTRACT

Melanoma pathogenesis from normal neural crest-derived melanocytes is often fatal due to aggressive cell invasion throughout the body. The identification of signals that reprogram de-differentiated, metastatic melanoma cells to a less aggressive and stable phenotype would provide a novel strategy to limit disease progression. In this study, we identify and test the function of developmental signals within the chick embryonic neural crest microenvironment to reprogram and sustain the transition of human metastatic melanoma to a neural crest cell-like phenotype. Results reveal that co-culture of the highly aggressive and metastatic human melanoma cell line C8161 upregulate a marker of melanosome formation (Mart-1) in the presence of embryonic day 3.5 chick trunk dorsal root ganglia. We identify nerve growth factor (NGF) as the signal within this tissue driving Mart-1 re-expression and show that NGF receptors trkA and p75 cooperate to induce Mart-1 re-expression. Furthermore, Mart-1 expressing C8161 cells acquire a gene signature of poorly aggressive C81-61 cells. These data suggest that targeting NGF signaling may yield a novel strategy to reprogram metastatic melanoma toward a benign cell type.

19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...