Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Annu Rev Cell Dev Biol ; 26: 581-603, 2010.
Article in English | MEDLINE | ID: mdl-19575671

ABSTRACT

The neural crest is a multipotent stem cell–like population that gives rise to a wide range of derivatives in the vertebrate embryo including elements of the craniofacial skeleton and peripheral nervous system as well as melanocytes. The neural crest forms in a series of regulatory steps that include induction and specification of the prospective neural crest territory–neural plate border, specification of bona fide neural crest progenitors, and differentiation into diverse derivatives. These individual processes during neural crest ontogeny are controlled by regulatory circuits that can be assembled into a hierarchical gene regulatory network (GRN). Here we present an overview of the GRN that orchestrates the formation of cranial neural crest cells. Formulation of this network relies on information largely inferred from gene perturbation studies performed in several vertebrate model organisms. Our representation of the cranial neural crest GRN also includes information about direct regulatory interactions obtained from the cis-regulatory analyses performed to date, which increases the resolution of the architectural circuitry within the network.


Subject(s)
Gene Regulatory Networks , Neural Crest/metabolism , Animals , Cell Movement , Gene Expression Regulation, Developmental , Neural Crest/cytology , Vertebrates/embryology
2.
Nat Rev Mol Cell Biol ; 9(7): 557-68, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18523435

ABSTRACT

The neural crest is a multipotent, migratory cell population that is unique to vertebrate embryos and gives rise to many derivatives, ranging from the peripheral nervous system to the craniofacial skeleton and pigment cells. A multimodule gene regulatory network mediates the complex process of neural crest formation, which involves the early induction and maintenance of the precursor pool, emigration of the neural crest progenitors from the neural tube via an epithelial to mesenchymal transition, migration of progenitor cells along distinct pathways and overt differentiation into diverse cell types. Here, we review our current understanding of these processes and discuss the molecular players that are involved in the neural crest gene regulatory network.


Subject(s)
Cell Differentiation/physiology , Gene Regulatory Networks , Neural Crest/embryology , Animals , Cell Cycle/physiology , Cell Movement/physiology , Embryonic Induction , Gap Junctions/metabolism , Gene Expression Regulation, Developmental , Morphogenesis , Neural Crest/anatomy & histology , Neural Crest/physiology , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/metabolism
3.
Genes Dev ; 25(21): 2306-20, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22056673

ABSTRACT

We report a multifunctional gene-trapping approach, which generates full-length Citrine fusions with endogenous proteins and conditional mutants from a single integration event of the FlipTrap vector. We identified 170 FlipTrap zebrafish lines with diverse tissue-specific expression patterns and distinct subcellular localizations of fusion proteins generated by the integration of an internal citrine exon. Cre-mediated conditional mutagenesis is enabled by heterotypic lox sites that delete Citrine and "flip" in its place mCherry with a polyadenylation signal, resulting in a truncated fusion protein. Inducing recombination with Cerulean-Cre results in fusion proteins that often mislocalize, exhibit mutant phenotypes, and dramatically knock down wild-type transcript levels. FRT sites in the vector enable targeted genetic manipulation of the trapped loci in the presence of Flp recombinase. Thus, the FlipTrap captures the functional proteome, enabling the visualization of full-length fluorescent fusion proteins and interrogation of function by conditional mutagenesis and targeted genetic manipulation.


Subject(s)
Proteome , Proteomics/methods , Animals , Bacterial Proteins/genetics , Databases, Protein , Embryo, Nonmammalian , Genetic Vectors , Internet , Luminescent Proteins/genetics , Molecular Sequence Annotation , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zebrafish
4.
Semin Cell Dev Biol ; 23(3): 320-32, 2012 May.
Article in English | MEDLINE | ID: mdl-22430756

ABSTRACT

Although the epithelial to mesenchymal transition (EMT) is famous for its role in cancer metastasis, it also is a normal developmental event in which epithelial cells are converted into migratory mesenchymal cells. A prime example of EMT during development occurs when neural crest (NC) cells emigrate from the neural tube thus providing an excellent model to study the principles of EMT in a nonmalignant environment. NC cells start life as neuroepithelial cells intermixed with precursors of the central nervous system. After EMT, they delaminate and begin migrating, often to distant sites in the embryo. While proliferating and maintaining multipotency and cell survival the transitioning neural crest cells lose apicobasal polarity and the basement membrane is broken down. This review discusses how these events are coordinated and regulated, by series of events involving signaling factors, gene regulatory interactions, as well as epigenetic and post-transcriptional modifications. Even though the series of events involved in NC EMT are well known, the sequence in which these steps take place remains a subject of debate, raising the intriguing possibility that, rather than being a single event, neural crest EMT may involve multiple parallel mechanisms.


Subject(s)
Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/physiology , Mesoderm/cytology , Neoplasms/pathology , Neural Crest/cytology , Animals , Cell Movement , Humans , Neural Crest/embryology
5.
Nature ; 453(7198): 1064-71, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18563158

ABSTRACT

Lancelets ('amphioxus') are the modern survivors of an ancient chordate lineage, with a fossil record dating back to the Cambrian period. Here we describe the structure and gene content of the highly polymorphic approximately 520-megabase genome of the Florida lancelet Branchiostoma floridae, and analyse it in the context of chordate evolution. Whole-genome comparisons illuminate the murky relationships among the three chordate groups (tunicates, lancelets and vertebrates), and allow not only reconstruction of the gene complement of the last common chordate ancestor but also partial reconstruction of its genomic organization, as well as a description of two genome-wide duplications and subsequent reorganizations in the vertebrate lineage. These genome-scale events shaped the vertebrate genome and provided additional genetic variation for exploitation during vertebrate evolution.


Subject(s)
Chordata/genetics , Evolution, Molecular , Genome/genetics , Animals , Chordata/classification , Conserved Sequence , DNA Transposable Elements/genetics , Gene Duplication , Genes/genetics , Genetic Linkage , Humans , Introns/genetics , Karyotyping , Multigene Family , Phylogeny , Polymorphism, Genetic/genetics , Proteins/genetics , Synteny , Time Factors , Vertebrates/classification , Vertebrates/genetics
6.
Development ; 137(3): 507-18, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081195

ABSTRACT

Comparative studies of the tetrapod raldh2 (aldh1a2) gene, which encodes a retinoic acid (RA) synthesis enzyme, have led to the identification of a dorsal spinal cord enhancer. Enhancer activity is directed dorsally to the roof plate and dorsal-most (dI1) interneurons through predicted Tcf- and Cdx-homeodomain binding sites and is repressed ventrally via predicted Tgif homeobox and ventral Lim-homeodomain binding sites. Raldh2 and Math1/Cath1 expression in mouse and chicken highlights a novel, transient, endogenous Raldh2 expression domain in dI1 interneurons, which give rise to ascending circuits and intraspinal commissural interneurons, suggesting roles for RA in the ontogeny of spinocerebellar and intraspinal proprioceptive circuits. Consistent with expression of raldh2 in the dorsal interneurons of tetrapods, we also found that raldh2 is expressed in dorsal interneurons throughout the agnathan spinal cord, suggesting ancestral roles for RA signaling in the ontogenesis of intraspinal proprioception.


Subject(s)
Aldehyde Oxidoreductases/physiology , Spinal Cord/physiology , Animals , Binding Sites , Chickens , Conserved Sequence , Evolution, Molecular , Hepatocyte Nuclear Factor 1-alpha , Homeodomain Proteins , Interneurons , LIM-Homeodomain Proteins , Mice , Mice, Transgenic , Repressor Proteins , T Cell Transcription Factor 1 , Transcription Factors , Tretinoin/physiology
7.
Nature ; 445(7128): 613-7, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-17237766

ABSTRACT

The organizer of the vertebrate gastrula is an important signalling centre that induces and patterns dorsal axial structures. Although a topic of long-standing interest, the evolutionary origin of the organizer remains unclear. Here we show that the gastrula of the cephalochordate amphioxus expresses dorsal/ventral (D/V) patterning genes (for example, bone morphogenetic proteins (BMPs), Nodal and their antagonists) in patterns reminiscent of those of their vertebrate orthlogues, and that amphioxus embryos, like those of vertebrates, are ventralized by exogenous BMP protein. In addition, Wnt-antagonists (for example, Dkks and sFRP2-like) are expressed anteriorly, whereas Wnt genes themselves are expressed posteriorly, consistent with a role for Wnt signalling in anterior/posterior (A/P) patterning. These results suggest evolutionary conservation of the mechanisms for both D/V and A/P patterning of the early gastrula. In light of recent phylogenetic analyses placing cephalochordates basally in the chordate lineage, we propose that separate signalling centres for patterning the D/V and A/P axes may be an ancestral chordate character.


Subject(s)
Biological Evolution , Body Patterning/physiology , Chordata/embryology , Organizers, Embryonic/physiology , Animals , Body Patterning/drug effects , Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/pharmacology , Chordata/genetics , Gastrula/metabolism , Gene Expression Regulation, Developmental , Organizers, Embryonic/drug effects , Signal Transduction , Transcription Factors/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 107(8): 3570-5, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20139305

ABSTRACT

The neural crest is a multipotent, stem cell-like population that migrates extensively in the embryo and forms a wide array of derivatives, ranging from neurons to melanocytes and cartilage. Analyses of the gene regulatory network driving neural crest development revealed Sox10 as one of the earliest neural crest-specifying genes, cell-autonomously driving delamination and directly regulating numerous downstream effectors and differentiation gene batteries. In search of direct inputs to the neural crest specifier module, we dissected the chick Sox10 genomic region and isolated two downstream regulatory regions with distinct spatiotemporal activity. A unique element, Sox10E2 represents the earliest-acting neural crest cis-regulatory element, critical for initiating Sox10 expression in newly formed cranial, but not vagal and trunk neural crest. A second element, Sox10E1, acts in later migrating vagal and trunk crest cells. Deep characterization of Sox10E2 reveals Sox9, Ets1, and cMyb as direct inputs mediating enhancer activity. ChIP, DNA-pull down, and gel-shift assays demonstrate their direct binding to the Sox10E2 enhancer in vivo, whereas mutation of their corresponding binding sites, or inactivation of the three upstream regulators, abolishes both reporter and endogenous Sox10 expression. Using cis-regulatory analysis as a tool, our study makes critical connections within the neural crest gene regulatory network, thus being unique in establishing a direct link of upstream effectors to a key neural crest specifier.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Neural Crest/embryology , SOXB1 Transcription Factors/metabolism , Skull/embryology , Animals , Base Sequence , Chick Embryo , Conserved Sequence , Genomics , Humans , Mice , Molecular Sequence Data , Multipotent Stem Cells/metabolism , Neural Crest/metabolism , Rats , Skull/metabolism , Transcriptional Activation , Xenopus
9.
Proc Natl Acad Sci U S A ; 107(40): 17262-7, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855630

ABSTRACT

The appearance of jaws was a turning point in vertebrate evolution because it allowed primitive vertebrates to capture and process large, motile prey. The vertebrate jaw consists of separate dorsal and ventral skeletal elements connected by a joint. How this structure evolved from the unjointed gill bar of a jawless ancestor is an unresolved question in vertebrate evolution. To understand the developmental bases of this evolutionary transition, we examined the expression of 12 genes involved in vertebrate pharyngeal patterning in the modern jawless fish lamprey. We find nested expression of Dlx genes, as well as combinatorial expression of Msx, Hand and Gsc genes along the dorso-ventral (DV) axis of the lamprey pharynx, indicating gnathostome-type pharyngeal patterning evolved before the appearance of the jaw. In addition, we find that Bapx and Gdf5/6/7, key regulators of joint formation in gnathostomes, are not expressed in the lamprey first arch, whereas Barx, which is absent from the intermediate first arch in gnathostomes, marks this domain in lamprey. Taken together, these data support a new scenario for jaw evolution in which incorporation of Bapx and Gdf5/6/7 into a preexisting DV patterning program drove the evolution of the jaw by altering the identity of intermediate first-arch chondrocytes. We present this "Pre-pattern/Cooption" model as an alternative to current models linking the evolution of the jaw to the de novo appearance of sophisticated pharyngeal DV patterning.


Subject(s)
Biological Evolution , Jaw/anatomy & histology , Lampreys , Models, Biological , Vertebrates/anatomy & histology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lampreys/anatomy & histology , Lampreys/genetics , Molecular Sequence Data , Transcription Factors/genetics , Transcription Factors/metabolism , Vertebrates/genetics
10.
Dev Cell ; 13(3): 405-20, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17765683

ABSTRACT

The vertebrate neural crest migrates from its origin, the neural plate border, to form diverse derivatives. We previously hypothesized that a neural crest gene regulatory network (NC-GRN) guides neural crest formation. Here, we investigate when during evolution this hypothetical network emerged by analyzing neural crest formation in lamprey, a basal extant vertebrate. We identify 50 NC-GRN homologs and use morpholinos to demonstrate a critical role for eight transcriptional regulators. The results reveal conservation in deployment of upstream factors, suggesting that proximal portions of the network arose early in vertebrate evolution and have been conserved for >500 million years. We found biphasic expression of neural crest specifiers and differences in deployment of some specifiers and effectors expected to confer species-specific properties. By testing the collective expression and function of neural crest genes in a single, basal vertebrate, we reveal the ground state of the NC-GRN and resolve ambiguities between model organisms.


Subject(s)
Evolution, Molecular , Gene Expression Regulation, Developmental , Neural Crest/embryology , Animals , DNA, Complementary , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Gene Library , In Situ Hybridization , Lampreys/embryology , Lampreys/genetics , Models, Biological , Oligonucleotides, Antisense/pharmacology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Development ; 136(24): 4155-64, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934013

ABSTRACT

Vertebrate cranial sensory ganglia have a dual origin from the neural crest and ectodermal placodes. In the largest of these, the trigeminal ganglion, Slit1-Robo2 signaling is essential for proper ganglion assembly. Here, we demonstrate a crucial role for the cell adhesion molecule N-cadherin and its interaction with Slit1-Robo2 during gangliogenesis in vivo. A common feature of chick trigeminal and epibranchial ganglia is the expression of N-cadherin and Robo2 on placodal neurons and Slit1 on neural crest cells. Interestingly, N-cadherin localizes to intercellular adherens junctions between placodal neurons during ganglion assembly. Depletion of N-cadherin causes loss of proper ganglion coalescence, similar to that observed after loss of Robo2, suggesting that the two pathways might intersect. Consistent with this possibility, blocking or augmenting Slit-Robo signaling modulates N-cadherin protein expression on the placodal cell surface concomitant with alteration in placodal adhesion. Lack of an apparent change in total N-cadherin mRNA or protein levels suggests post-translational regulation. Co-expression of N-cadherin with dominant-negative Robo abrogates the Robo2 loss-of-function phenotype of dispersed ganglia, whereas loss of N-cadherin reverses the aberrant aggregation induced by increased Slit-Robo expression. Our study suggests a novel mechanism whereby N-cadherin acts in concert with Slit-Robo signaling in mediating the placodal cell adhesion required for proper gangliogenesis.


Subject(s)
Cadherins/physiology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Sensory Receptor Cells/physiology , Trigeminal Ganglion/physiology , Animals , Cell Adhesion/physiology , Chick Embryo , Gene Expression Regulation, Developmental , Neural Crest/cytology , Neural Crest/physiology , Neurogenesis/physiology , Sensory Receptor Cells/cytology , Signal Transduction , Trigeminal Ganglion/cytology , Trigeminal Ganglion/embryology
12.
Nature ; 441(7094): 750-2, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16760978

ABSTRACT

The neural crest, a defining character of vertebrates, is of prime importance to their evolutionary origin. To understand neural crest evolution, we explored molecular mechanisms underlying craniofacial development in the basal jawless vertebrate, sea lamprey (Petromyzon marinus), focusing on the SoxE (Sox8, Sox9 and Sox10) gene family. In jawed vertebrates, these are important transcriptional regulators of the neural crest, and the loss of Sox9 causes abnormal craniofacial development. Here we report that two lamprey SoxE genes are expressed in migrating neural crest and crest-derived prechondrocytes in posterior branchial arches, whereas a third paralogue is expressed later in the perichondrium and mandibular arch. Morpholino knock-down of SoxE1 reveals that it is essential for posterior branchial arch development, although the mandibular arch is unaffected. The results show that chondrogenic function of SoxE regulators can be traced to the lamprey-gnathostome common ancestor and indicate that lamprey SoxE genes might have undergone independent duplication to have distinct functions in mandibular versus caudal branchial arches. This work sheds light on the homology of vertebrate branchial arches and supports their common origin at the base of vertebrates.


Subject(s)
Biological Evolution , Fish Proteins/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Petromyzon/embryology , Pharynx/embryology , Pharynx/metabolism , Animals , Branchial Region/cytology , Branchial Region/embryology , Branchial Region/metabolism , Fish Proteins/genetics , Molecular Sequence Data , Neural Crest/cytology , Petromyzon/genetics , Pharynx/cytology
13.
Nature ; 441(7090): 218-22, 2006 May 11.
Article in English | MEDLINE | ID: mdl-16688176

ABSTRACT

The neural crest is a stem population critical for development of the vertebrate craniofacial skeleton and peripheral ganglia. Neural crest cells originate along the border between the neural plate and epidermis, migrate extensively and generate numerous derivatives, including neurons and glia of the peripheral nervous system, melanocytes, bone and cartilage of the head skeleton. Impaired neural crest development is associated with human defects, including cleft palate. Classically, the neural crest has been thought to form by interactions at the border between neural and non-neural ectoderm or mesoderm, and defined factors such as bone morphogenetic proteins (BMPs) and Wnt proteins have been postulated as neural crest-inducers. Although competence to induce crest cells declines after stage 10 (ref. 14), little is known about when neural crest induction begins in vivo. Here we report that neural crest induction is underway during gastrulation and well before proper neural plate appearance. We show that a restricted region of chick epiblast (stage 3-4) is specified to generate neural crest cells when explanted under non-inducing conditions. This region expresses the transcription factor Pax7 by stage 4 + and later contributes to neural folds and migrating neural crest. In chicken embryos, Pax7 is required for neural crest formation in vivo, because blocking its translation inhibits expression of the neural crest markers Slug, Sox9, Sox10 and HNK-1. Our results indicate that neural crest specification initiates earlier than previously assumed, independently of mesodermal and neural tissues, and that Pax7 has a crucial function during neural crest development.


Subject(s)
Cell Differentiation , Gastrula/cytology , Gastrula/metabolism , Neural Crest/cytology , Neural Crest/embryology , PAX7 Transcription Factor/metabolism , Animals , Chick Embryo , Mesoderm/cytology , Mesoderm/metabolism , PAX7 Transcription Factor/genetics , Stem Cells/cytology , Stem Cells/metabolism
14.
Bioessays ; 32(9): 808-17, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20730948

ABSTRACT

Essentially we show recent data to shed new light on the thorny controversy of how teeth arose in evolution. Essentially we show (a) how teeth can form equally from any epithelium, be it endoderm, ectoderm or a combination of the two and (b) that the gene expression programs of oral versus pharyngeal teeth are remarkably similar. Classic theories suggest that (i) skin denticles evolved first and odontode-inductive surface ectoderm merged inside the oral cavity to form teeth (the 'outside-in' hypothesis) or that (ii) patterned odontodes evolved first from endoderm deep inside the pharyngeal cavity (the 'inside-out' hypothesis). We propose a new perspective that views odontodes as structures sharing a deep molecular homology, united by sets of co-expressed genes defining a competent thickened epithelium and a collaborative neural crest-derived ectomesenchyme. Simply put, odontodes develop 'inside and out', wherever and whenever these co-expressed gene sets signal to one another. Our perspective complements the classic theories and highlights an agenda for specific experimental manipulations in model and non-model organisms.


Subject(s)
Biological Evolution , Odontogenesis/genetics , Tooth/anatomy & histology , Vertebrates/anatomy & histology , Animals , Ectoderm/embryology , Ectoderm/physiology , Endoderm/embryology , Endoderm/physiology , Epithelium/embryology , Epithelium/physiology , Odontogenesis/physiology , Tooth/growth & development , Tooth/physiology , Tooth/ultrastructure , Vertebrates/genetics
15.
Nat Neurosci ; 11(3): 269-76, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18278043

ABSTRACT

Vertebrate cranial sensory ganglia, responsible for sensation of touch, taste and pain in the face and viscera, are composed of both ectodermal placode and neural crest cells. The cellular and molecular interactions allowing generation of complex ganglia remain unknown. Here, we show that proper formation of the trigeminal ganglion, the largest of the cranial ganglia, relies on reciprocal interactions between placode and neural crest cells in chick, as removal of either population resulted in severe defects. We demonstrate that ingressing placode cells express the Robo2 receptor and early migrating cranial neural crest cells express its cognate ligand Slit1. Perturbation of this receptor-ligand interaction by blocking Robo2 function or depleting either Robo2 or Slit1 using RNA interference disrupted proper ganglion formation. The resultant disorganization mimics the effects of neural crest ablation. Thus, our data reveal a novel and essential role for Robo2-Slit1 signaling in mediating neural crest-placode interactions during trigeminal gangliogenesis.


Subject(s)
Cell Movement/genetics , Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Crest/embryology , Receptors, Immunologic/metabolism , Stem Cells/metabolism , Trigeminal Ganglion/embryology , Animals , Cell Communication/genetics , Cell Differentiation/genetics , Chick Embryo , Chickens , Coturnix , Down-Regulation/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Neural Crest/cytology , Neural Crest/metabolism , RNA Interference , Receptors, Immunologic/genetics , Stem Cells/cytology , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism , Roundabout Proteins
16.
Dev Biol ; 340(2): 222-31, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-19913005

ABSTRACT

The transcription factor spalt4 is a key early-response gene in otic placode induction. Here, we characterize the cis-regulatory regions of spalt4 responsible for activation of its expression in the developing otic placode and report the isolation of a novel core enhancer. Identification and mutational analysis of putative transcription factor binding sites reveal that Pea3, a downstream effector of FGF signaling, and Pax2 directly activate spalt4 during ear development. Morpholino-mediated knock-down of each factor reduces or eliminates reporter expression. In contrast, combined over-expression of Pea3 and Pax2 drives ectopic reporter expression, suggesting that they function synergistically. These studies expand the gene regulatory network underlying early otic development by identifying direct inputs that mediate spalt4 expression.


Subject(s)
Ear/embryology , PAX2 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Chick Embryo , Embryo, Nonmammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Homeodomain Proteins , PAX2 Transcription Factor/genetics , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics
17.
Dev Biol ; 348(1): 107-18, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20883685

ABSTRACT

Glypicans are conserved cell surface heparan sulfate proteoglycans expressed in a spatiotemporally regulated manner in many developing tissues including the nervous system. Here, we show that Glypican-1 (GPC1) is expressed by trigeminal placode cells as they ingress and contribute to trigeminal sensory neurons in the chick embryo. Either expression of full-length or truncated GPC1 in vivo causes defects in trigeminal gangliogenesis in a manner that requires heparan sulfate side chains. This leads to either abnormal placodal differentiation or organization, respectively, with near complete loss of the ophthalmic (OpV) trigeminal ganglion in the most severe cases after overexpression of full-length GPC1. Interestingly, modulating GPC1 alters levels of endogenous Wnt signaling activity in the forming trigeminal ganglion, as indicated by Wnt reporter expression. Accordingly, GPC1 overexpression phenocopies Wnt inhibition in causing loss of OpV placodal neurons. Furthermore, increased Wnt activity rescues the effects of GPC1 overexpression. Taken together, these results suggest that appropriate levels of GPC1 are essential for proper regulation of canonical Wnt signaling during differentiation and organization of trigeminal placodal cells into ganglia.


Subject(s)
Gene Expression Regulation, Developmental , Glypicans/physiology , Nerve Tissue Proteins/physiology , Neurogenesis/physiology , Signal Transduction/physiology , Trigeminal Ganglion/embryology , Wnt Proteins/physiology , Animals , Chick Embryo , Glycosylphosphatidylinositols/metabolism , Glypicans/deficiency , Glypicans/genetics , Heparitin Sulfate/physiology , Phenotype , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/physiology , Sensory Receptor Cells/cytology , Trigeminal Ganglion/ultrastructure , beta Catenin/chemistry , beta Catenin/physiology
18.
Dev Cell ; 11(4): 505-17, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17011490

ABSTRACT

The sense organs of the vertebrate head comprise structures as varied as the eye, inner ear, and olfactory epithelium. In the early embryo, these assorted structures share a common developmental origin within the preplacodal region and acquire specific characteristics only later. Here we demonstrate a fundamental similarity in placodal precursors: in the chick all are specified as lens prior to acquiring features of specific sensory or neurogenic placodes. Lens specification becomes progressively restricted in the head ectoderm, initially by FGF and subsequently by signals derived from migrating neural crest cells. We show that FGF8 from the anterior neural ridge is both necessary and sufficient to promote olfactory fate in adjacent ectoderm. Our results reveal that placode precursors share a common ground state as lens and progressive restriction allows the full range of placodal derivatives to form.


Subject(s)
Embryonic Induction , Fibroblast Growth Factors/metabolism , Lens, Crystalline/embryology , Sense Organs/embryology , Animals , Chick Embryo , Fibroblast Growth Factors/genetics , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/cytology , Models, Biological , Organ Culture Techniques
19.
Proc Natl Acad Sci U S A ; 105(51): 20083-8, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19104059

ABSTRACT

The neural crest, a multipotent embryonic cell type, originates at the border between neural and nonneural ectoderm. After neural tube closure, these cells undergo an epithelial-mesenchymal transition, migrate to precise, often distant locations, and differentiate into diverse derivatives. Analyses of expression and function of signaling and transcription factors in higher vertebrates has led to the proposal that a neural crest gene regulatory network (NC-GRN) orchestrates neural crest formation. Here, we interrogate the NC-GRN in the lamprey, taking advantage of its slow development and basal phylogenetic position to resolve early inductive events, 1 regulatory step at the time. To establish regulatory relationships at the neural plate border, we assess relative expression of 6 neural crest network genes and effects of individually perturbing each on the remaining 5. The results refine an upstream portion of the NC-GRN and reveal unexpected order and linkages therein; e.g., lamprey AP-2 appears to function early as a neural plate border rather than a neural crest specifier and in a pathway linked to MsxA but independent of ZicA. These findings provide an ancestral framework for performing comparative tests in higher vertebrates in which network linkages may be more difficult to resolve because of their rapid development.


Subject(s)
Gene Regulatory Networks , Neural Crest/growth & development , Petromyzon/embryology , Animals , Biological Evolution , Neural Crest/cytology , Neural Plate , Systems Biology/methods , Transcription Factor AP-2
20.
Dev Biol ; 336(2): 257-65, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19833121

ABSTRACT

Cranial neural crest cells migrate into the periocular region and later contribute to various ocular tissues including the cornea, ciliary body and iris. After reaching the eye, they initially pause before migrating over the lens to form the cornea. Interestingly, removal of the lens leads to premature invasion and abnormal differentiation of the cornea. In exploring the molecular mechanisms underlying this effect, we find that semaphorin3A (Sema3A) is expressed in the lens placode and epithelium continuously throughout eye development. Interestingly, neuropilin-1 (Npn-1) is expressed by periocular neural crest but down-regulated, in a manner independent of the lens, by the subpopulation that migrates into the eye and gives rise to the cornea endothelium and stroma. In contrast, Npn-1 expressing neural crest cells remain in the periocular region and contribute to the anterior uvea and ocular blood vessels. Introduction of a peptide that inhibits Sema3A/Npn-1 signaling results in premature entry of neural crest cells over the lens that phenocopies lens ablation. Furthermore, Sema3A inhibits periocular neural crest migration in vitro. Taken together, our data reveal a novel and essential role of Sema3A/Npn-1 signaling in coordinating periocular neural crest migration that is vital for proper ocular development.


Subject(s)
Cornea/embryology , Neural Crest/embryology , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Signal Transduction , Animals , Chick Embryo , Coturnix , Immunohistochemistry , In Situ Hybridization
SELECTION OF CITATIONS
SEARCH DETAIL