Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (184)2022 06 16.
Article in English | MEDLINE | ID: mdl-35786636

ABSTRACT

The inner ear perceives sound and maintains balance using the cochlea and vestibule. It does this by using a dedicated mechanosensory cell type known as the hair cell. Basic research in the inner ear has led to a deep understanding of how the hair cell functions, and how dysregulation can lead to hearing loss and vertigo. For this research, the mouse has been the pre-eminent model system. However, mice, like all mammals, have lost the ability to replace hair cells. Thus, when trying to understand cellular therapies for restoring inner ear function, complementary studies in other vertebrate species could provide further insights. The auditory epithelium of birds, the basilar papilla (BP), is a sheet of epithelium composed of mechanosensory hair cells (HCs) intercalated by supporting cells (SCs). Although the anatomical architecture of the basilar papilla and the mammalian cochlea differ, the molecular mechanisms of inner ear development and hearing are similar. This makes the basilar papilla a useful system for not only comparative studies but also to understand regeneration. Here, we describe dissection and manipulation techniques for the chicken inner ear. The technique shows genetic and small molecule inhibition methods, which offer a potent tool for studying the molecular mechanisms of inner ear development. In this paper, we discuss in ovo electroporation techniques to genetically perturb the basilar papilla using CRIPSR-Cas9 deletions, followed by dissection of the basilar papilla. We also demonstrate the BP organ culture and optimal use of culture matrices, to observe the development of the epithelium and the hair cells.


Subject(s)
Organ of Corti , Vestibule, Labyrinth , Animals , Chickens , Cochlea , Hair Cells, Auditory , Mammals , Mice
2.
Proc Natl Acad Sci U S A ; 115(33): 8388-8393, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061390

ABSTRACT

The mechanosensory hair cells of the inner ear are required for hearing and balance and have a distinctive apical structure, the hair bundle, that converts mechanical stimuli into electrical signals. This structure comprises a single cilium, the kinocilium, lying adjacent to an ensemble of actin-based projections known as stereocilia. Hair bundle polarity depends on kinociliary protocadherin-15 (Pcdh15) localization. Protocadherin-15 is found only in hair-cell kinocilia, and is not localized to the primary cilia of adjacent supporting cells. Thus, Pcdh15 must be specifically targeted and trafficked into the hair-cell kinocilium. Here we show that kinocilial Pcdh15 trafficking relies on cell type-specific coupling to the generic intraflagellar transport (IFT) transport mechanism. We uncover a role for fibroblast growth factor receptor 1 (FGFR1) in loading Pcdh15 onto kinociliary transport particles in hair cells. We find that on activation, FGFR1 binds and phosphorylates Pcdh15. Moreover, we find a previously uncharacterized role for clathrin in coupling this kinocilia-specific cargo with the anterograde IFT-B complex through the adaptor, DAB2. Our results identify a modified ciliary transport pathway used for Pcdh15 transport into the cilium of the inner ear hair cell and coordinated by FGFR1 activity.


Subject(s)
Cadherins/physiology , Flagella/metabolism , Hair Cells, Auditory, Inner/metabolism , Protein Precursors/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/physiology , Animals , Apoptosis Regulatory Proteins , Cadherin Related Proteins , Chick Embryo , Clathrin/physiology , Mice , Phosphorylation , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/analysis
3.
Semin Cell Dev Biol ; 65: 39-46, 2017 05.
Article in English | MEDLINE | ID: mdl-27989562

ABSTRACT

The inner ear arises from non-neural ectoderm as a result of instructions sent by surrounding tissues. These interactions progressively restrict the potential of the ectoderm, resulting in the formation of the otic placode, a disk of thickened ectoderm that will give rise to all of the inner ear derivatives and its neurons. While otic placode is a surface structure, the inner ear is internalised, embedded within the cranial mesenchyme. Here, the cellular and molecular interactions that restrict the lineage of non-neural ectoderm in its transition to otic placode are reviewed, and how these interactions impinge on the coordination of otic placodal cell shape that drive the dramatic morphogenesis of the placode, as it becomes the otocyst.


Subject(s)
Cell Lineage/genetics , Ear, Inner/growth & development , Ear, Inner/metabolism , Ectoderm/metabolism , Gene Expression Regulation, Developmental , Animals , Chick Embryo , Ear, Inner/cytology , Ectoderm/cytology , Embryo, Mammalian , Embryo, Nonmammalian , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fishes/growth & development , Fishes/metabolism , Mice , Organogenesis/genetics , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus/growth & development , Xenopus/metabolism
4.
Elife ; 4: e07178, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26359635

ABSTRACT

Innate pluripotency of mouse embryos transits from naive to primed state as the inner cell mass differentiates into epiblast. In vitro, their counterparts are embryonic (ESCs) and epiblast stem cells (EpiSCs), respectively. Activation of the FGF signaling cascade results in mouse ESCs differentiating into mEpiSCs, indicative of its requirement in the shift between these states. However, only mouse ESCs correspond to the naive state; ESCs from other mammals and from chick show primed state characteristics. Thus, the significance of the naive state is unclear. In this study, we use zebra finch as a model for comparative ESC studies. The finch blastoderm has mESC-like properties, while chick blastoderm exhibits EpiSC features. In the absence of FGF signaling, finch cells retained expression of pluripotent markers, which were lost in cells from chick or aged finch epiblasts. Our data suggest that the naive state of pluripotency is evolutionarily conserved among amniotes.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Finches/embryology , Germ Layers/cytology , Germ Layers/physiology , Animals , Biomarkers/analysis , Finches/growth & development
5.
Genesis ; 53(11): 669-77, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26385755

ABSTRACT

The domesticated zebra finch (Taeniopygia guttata) is a well-established animal model for studying vocal learning. It is also a tractable model for developmental analyses. The finch genome has been sequenced and methods for its transgenesis have been reported. Hatching and sexual maturation in this species takes only two weeks and three months, respectively. Finch colonies can be established relatively easily and its eggs are laid at a stage earlier than in other common avian experimental models, facilitating the analysis of very early avian development. Representing the Neoaves to which 95% of all bird species belong, the finch can potentially complement two existing, Galloanserae developmental models, the chick, and quail. Here, we provide a step-by-step guide for how to set up a finch colony in a conventional laboratory environment. Technical tips are offered to optimize hens' productivity and ensure a constant supply of fertilized finch eggs. Methods of handling finch eggs and embryos for subsequent embryological, cellular, or molecular analyses are also discussed. We conclude by emphasizing scientific values and cost effectiveness of maintaining a finch colony for avian developmental studies. genesis 53:669-677, 2015. © 2015 Wiley Periodicals, Inc.


Subject(s)
Developmental Biology/methods , Finches/growth & development , Models, Biological , Animal Husbandry , Animals , Female , Housing, Animal , Male , Photoperiod , Reproduction , Sex Determination Analysis
6.
Front Pharmacol ; 6: 19, 2015.
Article in English | MEDLINE | ID: mdl-25713536

ABSTRACT

Various cellular replacement therapies using in vitro generated cells to replace damaged tissue have been proposed as strategies to alleviate hearing loss. All such therapies must involve a complete understanding of the earliest steps in inner ear development; its induction as a thickened plate of cells in the non-neural, surface ectoderm of the embryo, to its internalization as an otocyst embedded in the head mesenchyme of the embryo. Such knowledge informs researchers addressing the feasibility of the proposed strategy and present alternatives if needed. In this review we describe the mechanisms of inner ear induction, concentrating on the factors that steer the fate of ectoderm into precursors of the inner ear. Induction then leads to inner ear morphogenesis and we describe the cellular changes that occur as the inner ear is converted from a superficial placode to an internalized otocyst, and how they are coordinated with a particular emphasis on how the signaling environment surrounding the inner ear influences these processes.

7.
Biol Cell ; 107(2): 41-60, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25412697

ABSTRACT

BACKGROUND INFORMATION: The vertebrate basic helix-loop-helix transcription factor Atoh1 is essential for maturation and survival of mechanosensory hair cells of the inner ear, neurogenesis, differentiation of the intestine, homeostasis of the colon and is implicated in cancer progression. Given that mutations in Atoh1 are detected in malignant tumours, study of functionally different Atoh1 alleles and homologues might yield useful avenues for investigation. The predicted sequence of chicken Atoh1 (cAtoh1) has large regions of dissimilarity to that of mammalian Atoh1 homologues. We hypothesise that cAtoh1 might have intrinsic functional differences to mammalian Atoh1. RESULTS: In this study, we cloned and sequenced the full open reading frame of cAtoh1. In overexpression experiments, we show that this sequence is sufficient to generate a cAtoh1 protein capable of inducing hair cell markers when expressed in nonsensory regions of the developing inner ear, and that morpholino-mediated knock-down using a section of the sequence 5' to the start codon inhibits differentiation of hair cells in the chicken basilar papilla. Furthermore, we compare the behaviour of cAtoh1 and human Atoh1 (hAtoh1) in embryonic mouse cochlear explants, showing that cAtoh1 is a potent inducer of hair cell differentiation and that it can overcome Sox2-mediated repression of hair cell differentiation more effectively than hAtoh1. CONCLUSIONS: cAtoh1 is both necessary and sufficient for avian mechanosensory hair cell differentiation. The non-conserved regions of the cAtoh1 coding region have functional consequences on its behaviour.


Subject(s)
Avian Proteins/genetics , Avian Proteins/metabolism , Chickens/genetics , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Base Sequence , Biomarkers/metabolism , Cell Differentiation , Cloning, Molecular , Cochlea/metabolism , Gene Knockdown Techniques , HEK293 Cells , Hair Cells, Auditory, Inner/cytology , Hair Cells, Auditory, Inner/metabolism , Humans , Labyrinth Supporting Cells/metabolism , Mammals/metabolism , Mice , Molecular Sequence Data , Molecular Weight , SOXB1 Transcription Factors/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
8.
Dev Dyn ; 244(2): 168-80, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25370455

ABSTRACT

BACKGROUND: Inner ear morphogenesis is tightly regulated by the temporally and spatially coordinated action of signaling ligands and their receptors. Ligand-receptor interactions are influenced by heparan sulfate proteoglycans (HSPGs), cell surface molecules that consist of glycosaminoglycan chains bound to a protein core. Diversity in the sulfation pattern within glycosaminoglycan chains creates binding sites for numerous cell signaling factors, whose activities and distribution are modified by their association with HSPGs. RESULTS: Here we describe the expression patterns of two extracellular 6-O-endosulfatases, Sulf1 and Sulf2, whose activity modifies the 6-O-sulfation pattern of HSPGs. We use in situ hybridization to determine the temporal and spatial distribution of transcripts during the development of the chick and mouse inner ear. We also use immunocytochemistry to determine the cellular localization of Sulf1 and Sulf2 within the sensory epithelia. Furthermore, we analyze the organ of Corti in Sulf1/Sulf2 double knockout mice and describe an increase in the number of mechanosensory hair cells. CONCLUSIONS: Our results suggest that the tuning of intracellular signaling, mediated by Sulf activity, plays an important role in the development of the inner ear.


Subject(s)
Avian Proteins/biosynthesis , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Organ of Corti/embryology , Sulfatases/biosynthesis , Sulfotransferases/biosynthesis , Animals , Chick Embryo , Mice , Organ of Corti/cytology , Signal Transduction/physiology
9.
Dev Biol ; 394(2): 206-16, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25173873

ABSTRACT

After induction, the inner ear is transformed from a superficially located otic placode into an epithelial vesicle embedded in the mesenchyme of the head. Invagination of this epithelium is biphasic: phase 1 involves the expansion of the basal aspect of the otic cells, and phase 2, the constriction of their apices. Apical constriction is important not only for otic invagination, but also the invagination of many other epithelia; however, its molecular basis is still poorly understood. Here we show that phase 2 otic morphogenesis, like phase 1 morphogenesis, results from the activation of myosin-II. However unlike the actin depolymerising activity observed basally, active myosin-II results in actomyosin contractility. Myosin-II activation is triggered by the accumulation of the planar cell polarity (PCP) core protein, Celsr1 in apical junctions (AJ). Apically polarized Celsr1 orients and recruits the Rho Guanine exchange factor (GEF) ArhGEF11 to apical junctions, thus restricting RhoA activity to the junctional membrane where it activates the Rho kinase ROCK. We suggest that myosin-II and RhoA activation results in actomyosin dependent constriction in an apically polarised manner driving otic epithelium invagination.


Subject(s)
Ear, Inner/embryology , Gene Expression Regulation, Developmental/physiology , Morphogenesis/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Azepines , Blotting, Western , Cadherins/metabolism , Chick Embryo , Ear, Inner/metabolism , Electroporation , Heterocyclic Compounds, 4 or More Rings , Image Processing, Computer-Assisted , Immunohistochemistry , Naphthalenes , Peptides , RNA Interference , rho-Associated Kinases/metabolism
10.
PLoS Genet ; 10(1): e1004118, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24465223

ABSTRACT

Inner ear mechanosensory hair cells transduce sound and balance information. Auditory hair cells emerge from a Sox2-positive sensory patch in the inner ear epithelium, which is progressively restricted during development. This restriction depends on the action of signaling molecules. Fibroblast growth factor (FGF) signalling is important during sensory specification: attenuation of Fgfr1 disrupts cochlear hair cell formation; however, the underlying mechanisms remain unknown. Here we report that in the absence of FGFR1 signaling, the expression of Sox2 within the sensory patch is not maintained. Despite the down-regulation of the prosensory domain markers, p27(Kip1), Hey2, and Hes5, progenitors can still exit the cell cycle to form the zone of non-proliferating cells (ZNPC), however the number of cells that form sensory cells is reduced. Analysis of a mutant Fgfr1 allele, unable to bind to the adaptor protein, Frs2/3, indicates that Sox2 maintenance can be regulated by MAP kinase. We suggest that FGF signaling, through the activation of MAP kinase, is necessary for the maintenance of sensory progenitors and commits precursors to sensory cell differentiation in the mammalian cochlea.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Ear, Inner/growth & development , Hair Cells, Auditory, Inner/cytology , Membrane Proteins/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle , Cell Differentiation/genetics , Cochlea/growth & development , Cochlea/metabolism , Ear, Inner/cytology , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 1/metabolism , SOXB1 Transcription Factors/genetics , Signal Transduction
11.
Methods ; 66(3): 447-53, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-23792918

ABSTRACT

The inner ear transduces the mechanical stimuli that are associated with sound and balance perception. Missteps during its formation often result in deafness, and thus understanding otic development has a profound clinical relevance. The intricate complexity of the inner ear is derived from a simple epithelial sheet during embryogenesis. Study of this process in vitro has provided insight into the mechanisms of otic induction, patterning and differentiation. This article details methods for the culture of otic placode, otocyst, and basilar papilla, providing a toolkit for the investigation of multiple facets of otic organogenesis, for regeneration studies and for setting up small molecule screens to identify possible therapeutic targets.


Subject(s)
Chickens , Ear, Inner/embryology , Tissue Culture Techniques , Animals , Chick Embryo
12.
Neural Dev ; 8: 13, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23829703

ABSTRACT

BACKGROUND: In order to fulfill their chemosensory function, olfactory neurons are in direct contact with the external environment and are therefore exposed to environmental aggressive factors. Olfaction is maintained through life because, unlike for other sensory neuroepithelia, olfactory neurons have a unique capacity to regenerate after trauma. The mechanisms that control the ontogenesis and regenerative ability of these neurons are not fully understood. Here, we used various experimental approaches in two model systems (chick and mouse) to assess the contribution of retinoic acid signaling in the induction of the olfactory epithelium, the generation and maintenance of progenitor populations, and the ontogenesis and differentiation of olfactory neurons. RESULTS: We show that retinoic acid signaling, although dispensable for initial induction of the olfactory placode, plays a key role in neurogenesis within this neuroepithelium. Retinoic acid depletion in the olfactory epithelium, both in chick and mouse models, results in a failure of progenitor cell maintenance and, consequently, differentiation of olfactory neurons is not sustained. Using an explant system, we further show that renewal of olfactory neurons is hindered if the olfactory epithelium is unable to synthesize retinoic acid. CONCLUSIONS: Our data show that retinoic acid is not a simple placodal inductive signal, but rather controls olfactory neuronal production by regulating the fate of olfactory progenitor cells. Retinaldehyde dehydrogenase 3 (RALDH3) is the key enzyme required to generate retinoic acid within the olfactory epithelium.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Neurogenesis/drug effects , Olfactory Bulb/drug effects , Olfactory Receptor Neurons/drug effects , Stem Cells/drug effects , Tretinoin/pharmacology , Animals , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Mice , Neurogenesis/physiology , Olfactory Bulb/cytology , Olfactory Mucosa/cytology , Olfactory Mucosa/drug effects , Olfactory Receptor Neurons/cytology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/cytology
13.
Nat Commun ; 3: 1041, 2012.
Article in English | MEDLINE | ID: mdl-22948823

ABSTRACT

The paratympanic organ, a mechanosensory hair cell-containing pouch in the amniote middle ear, was first described 100 years ago, yet its origins remain unresolved. Homology with the anamniote spiracular organ is supported by association with homologous skeletal elements and similar central targets of afferent neurons, suggesting it might be a remnant of the water-dependent lateral line system, otherwise lost during the amniote transition to terrestrial life. However, this is incompatible with studies suggesting that it arises from the first epibranchial (geniculate) placode. Here we show that a previously undiscovered Sox2-positive placode, immediately dorsal to the geniculate placode, forms the paratympanic organ and its afferent neurons, which are molecularly and morphologically distinct from geniculate neurons. These data remove the only obstacle to accepting the homology of the paratympanic organ and spiracular organ. We hypothesize that the paratympanic organ/spiracular organ represents an ancient head ectoderm module, developmentally and evolutionarily independent of both lateral line and epibranchial placodes.


Subject(s)
Ear, Middle/embryology , Ectoderm/embryology , Hair Cells, Auditory/cytology , Vertebrates/embryology , Animals , Biological Evolution , Chick Embryo , Chickens/metabolism , Ear, Middle/cytology , Ear, Middle/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Hair Cells, Auditory/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Phylogeny , Quail/embryology , Quail/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sense Organs/embryology , Sense Organs/metabolism , Sharks/embryology , Sharks/metabolism , Vertebrates/classification , Vertebrates/genetics , Vertebrates/metabolism
14.
Dev Dyn ; 241(11): 1716-28, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22972769

ABSTRACT

BACKGROUND: The inner ear and epibranchial ganglia of vertebrates arise from a shared progenitor domain that is induced by FGF signalling, the posterior placodal area (PPA), before being segregated by Wnt signalling. One of the first genes activated in the PPA is the transcription factor Pax2. Loss-of- and gain-of function studies have defined a role for Pax2 in placodal morphogenesis and later inner ear development, but have not addressed the role Pax2 plays during the formation and maintenance of the PPA. RESULTS: To understand the role of Pax2 during the development of the PPA, we used over-expression and repression of Pax2. Both gave rise to a smaller otocyst and repressed the formation of epibranchial placodes. In addition, cell cycle analysis revealed that Pax2 suppression reduced proliferation of the PPA. CONCLUSIONS: Our results suggest that Pax2 functions in the maintenance but not the induction of the PPA. One role of Pax2 is to maintain proper cell cycle proliferation in the PPA.


Subject(s)
PAX2 Transcription Factor/metabolism , Animals , Chick Embryo , Electroporation , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , In Situ Hybridization , PAX2 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism
15.
Dev Dyn ; 241(6): 1104-10, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22473893

ABSTRACT

BACKGROUND: The auditory complex of the chick, like that of humans, is made of intimate and highly ordered connections between the inner ear, the middle ear, and the outer ear. Unlike mammals, the middle ear of chick has only one ossicle, known as the columella. The independent lineages of the two suggest that some mechanism must exist that ensures the connectivity between the inner ear and the columella; however, the basis of integration is not known. RESULTS: Using quail-chick chimeras, we demonstrate that columella development depends on signaling interactions. Specifically, both pharyngeal endoderm and cranial paraxial mesoderm can alter the morphology of the columella. Only a discrete region of pharyngeal endoderm exerts this patterning activity, and this region is specified by the overlying paraxial mesoderm. CONCLUSIONS: Paraxial mesoderm is also used in the induction of the inner ear, thus we propose that this overlapping source of signalling cues in both middle and inner ear development may underlie the integration of these structures.


Subject(s)
Ear Ossicles/embryology , Ear, Inner/embryology , Embryonic Induction/physiology , Endoderm/physiology , Mesoderm/physiology , Morphogenesis/physiology , Signal Transduction/physiology , Alcian Blue , Animals , Chick Embryo , Chimera/embryology , Immunohistochemistry , Quail
16.
Dev Cell ; 21(4): 607-8, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-22014519

ABSTRACT

The earliest steps in tooth development depend on signaling interactions that result in the condensation of mandibular mesenchyme into the tooth bud. Reporting in this issue of Developmental Cell, Mammoto et al. (2011) find that chemotactic signals coordinate condensation and that the compressive force generated is sufficient to induce tooth bud gene expression.

17.
Development ; 137(11): 1777-85, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20460364

ABSTRACT

The inner ear and the epibranchial ganglia constitute much of the sensory system in the caudal vertebrate head. The inner ear consists of mechanosensory hair cells, their neurons, and structures necessary for sound and balance sensation. The epibranchial ganglia are knots of neurons that innervate and relay sensory signals from several visceral organs and the taste buds. Their development was once thought to be independent, in line with their independent functions. However, recent studies indicate that both systems arise from a morphologically distinct common precursor domain: the posterior placodal area. This review summarises recent studies into the induction, morphogenesis and innervation of these systems and discusses lineage restriction and cell specification in the context of their common origin.


Subject(s)
Ear, Inner/embryology , Ear, Inner/innervation , Ganglia, Sensory/embryology , Animals , Body Patterning , Branchial Region/embryology , Branchial Region/innervation , Chick Embryo , Embryonic Induction , Fibroblast Growth Factors/physiology , Lateral Line System/embryology , Lateral Line System/innervation , Mice , Models, Biological , Neurogenesis , Signal Transduction , Zebrafish/embryology
18.
Dev Biol ; 330(2): 389-98, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19362544

ABSTRACT

The eye field is initially a large single domain at the anterior end of the neural plate and is the first indication of optic potential in the vertebrate embryo. During the course of development, this domain is subject to interactions that shape and refine the organogenic field. The action of the prechordal mesoderm in bisecting this single region into two bilateral domains has been well described, however the role of signalling interactions in the further restriction and refinement of this domain has not been previously characterised. Here we describe a role for the rostral cephalic paraxial mesoderm in limiting the extent of the eye field. The anterior transposition of this mesoderm or its ablation disrupted normal development of the eye. Importantly, perturbation of optic vesicle development occurred in the absence of any detectable changes in the pattern of neighbouring regions of the neural tube. Furthermore, negative regulation of eye development is a property unique to the rostral paraxial mesoderm. The rostral paraxial mesoderm expresses members of the bone morphogenetic protein (BMP) family of signalling molecules and manipulation of endogenous BMP signalling resulted in abnormalities of the early optic primordia.


Subject(s)
Bone Morphogenetic Proteins/physiology , Eye/embryology , Mesoderm/embryology , Animals , Chick Embryo , Coturnix/embryology , Electroporation , In Situ Hybridization , Recombinant Proteins/metabolism
19.
Development ; 135(20): 3415-24, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18799542

ABSTRACT

The development of the vertebrate inner ear is an emergent process. Its progression from a relatively simple disk of thickened epithelium within head ectoderm into a complex organ capable of sensing sound and balance is controlled by sequential molecular and cellular interactions. Fibroblast growth factor (FGF) and Wnt signals emanating from mesoderm and neural ectoderm have been shown to direct inner ear fate. However, the role of these multiple signals during inner ear induction is unclear. We demonstrate that the action of the FGFs and Wnts is sequential, and that their roles support a model of hierarchical fate decisions that progressively restrict the developmental potential of the ectoderm until otic commitment. We show that signalling by Fgf3 and Fgf19 is required to initiate a proliferative progenitor region that is a precursor to both the inner ear and the neurogenic epibranchial placodes. Significantly, we find that only after FGF action is attenuated can the subsequent action of Wnt signalling allow otic differentiation to proceed. In addition, gain and loss of function of Wnt-signalling components show a role for this signalling in repressing epibranchial fate. This interplay of signalling factors ensures the correct and ordered differentiation of both inner ear and epibranchial systems.


Subject(s)
Ear, Inner/embryology , Ear, Inner/physiology , Fibroblast Growth Factors/physiology , Wnt Proteins/physiology , Animals , Chick Embryo , Ear, Inner/metabolism , Embryo, Nonmammalian , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Models, Biological , Wnt Proteins/genetics , Wnt Proteins/metabolism
20.
Curr Biol ; 18(13): 976-81, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18583133

ABSTRACT

Changes in the cytoskeletal architecture underpin the dynamic changes in tissue shape that occur during development. It is clear that such changes must be coordinated so that individual cell behaviors are synchronized; however, the mechanisms by which morphogenesis is instructed and coordinated are unknown. After its induction in non-neural ectoderm, the inner ear undergoes morphogenesis, being transformed from a flat ectodermal disk on the surface of the embryo to a hollowed sphere embedded in the head. We provide evidence that this shape change relies on extrinsic signals subsequent to genetic specification. By using specific inhibitors, we find that local fibroblast growth factor (FGF) signaling triggers a phosphorylation cascade that activates basal myosin II through the activation of phospholipase Cgamma. Myosin II exhibits a noncanonical activity that results in the local depletion of actin filaments. Significantly, the resulting apical actin enrichment drives morphogenesis of the inner ear. Thus, FGF signaling directly exerts profound cytoskeletal effects on otic cells, coordinating the morphogenesis of the inner ear. The iteration of this morphogenetic signaling system suggests that it is a more generally applicable mechanism in other epithelial tissues undergoing shape change.


Subject(s)
Cytoskeleton/physiology , Ear, Inner/embryology , Epithelium/embryology , Fibroblast Growth Factors/metabolism , Morphogenesis , Actins/metabolism , Animals , Chick Embryo , Ear, Inner/metabolism , Ectoderm/metabolism , Enzyme Activation , Myosin Type II/metabolism , Phospholipase C gamma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...