Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Development ; 150(21)2023 Nov 01.
Article En | MEDLINE | ID: mdl-37938828

Cranial ganglia are aggregates of sensory neurons that mediate distinct types of sensation. The statoacoustic ganglion (SAG) develops into several lobes that are spatially arranged to connect appropriately with hair cells of the inner ear. To investigate the cellular behaviours involved in the 3D organization of the SAG, we use high-resolution confocal imaging of single-cell, labelled zebrafish neuroblasts (NBs), photoconversion, photoablation, and genetic perturbations. We show that otic NBs delaminate out of the otic epithelium in an epithelial-mesenchymal transition-like manner, rearranging apical polarity and primary cilia proteins. We also show that, once delaminated, NBs require RhoGTPases in order to perform active migration. Furthermore, tracking of recently delaminated NBs revealed their directed migration and coalescence around a small population of pioneer SAG neurons. These pioneer SAG neurons, not from otic placode origin, populate the coalescence region before otic neurogenesis begins and their ablation disrupts delaminated NB migratory pathways, consequentially affecting SAG shape. Altogether, this work shows for the first time the role of pioneer SAG neurons in orchestrating SAG development.


Ear, Inner , Zebrafish , Animals , Zebrafish/genetics , Cell Differentiation/genetics , Ear, Inner/metabolism , Hair Cells, Auditory/physiology , Sensory Receptor Cells
2.
Dev Dyn ; 252(1): 81-103, 2023 01.
Article En | MEDLINE | ID: mdl-35972036

Sensory neurons of the head are the ones that transmit the information about the external world to our brain for its processing. Axons from cranial sensory neurons sense different chemoattractant and chemorepulsive molecules during the journey and in the target tissue to establish the precise innervation with brain neurons and/or receptor cells. Here, we aim to unify and summarize the available information regarding molecular mechanisms guiding the different afferent sensory axons of the head. By putting the information together, we find the use of similar guidance cues in different sensory systems but in distinct combinations. In vertebrates, the number of genes in each family of guidance cues has suffered a great expansion in the genome, providing redundancy, and robustness. We also discuss recently published data involving the role of glia and mechanical forces in shaping the axon paths. Finally, we highlight the remaining questions to be addressed in the field.


Axon Guidance , Axons , Animals , Axons/physiology , Sensory Receptor Cells , Neuroglia , Sense Organs
4.
Cell Rep ; 37(7): 110004, 2021 11 16.
Article En | MEDLINE | ID: mdl-34788624

Polyphosphate (polyP) is a polymer of hundreds of phosphate residues present in all organisms. In mammals, polyP is involved in crucial physiological processes, including coagulation, inflammation, and stress response. However, after decades of research, the metabolic enzymes are still unknown. Here, we purify and identify Nudt3, a NUDIX family member, as the enzyme responsible for polyP phosphatase activity in mammalian cells. We show that Nudt3 shifts its substrate specificity depending on the cation; specifically, Nudt3 is active on polyP when Zn2+ is present. Nudt3 has in vivo polyP phosphatase activity in human cells, and importantly, we show that cells with altered polyP levels by modifying Nudt3 protein amount present reduced viability upon oxidative stress and increased DNA damage, suggesting that polyP and Nudt3 play a role in oxidative stress protection. Finally, we show that Nudt3 is involved in the early stages of embryo development in zebrafish.


Acid Anhydride Hydrolases/metabolism , Oxidative Stress/physiology , Polyphosphates/metabolism , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/physiology , Animals , HEK293 Cells , Humans , Male , Mammals/metabolism , Oxidation-Reduction , Phosphoric Monoester Hydrolases/physiology , Rats , Rats, Sprague-Dawley , Substrate Specificity/physiology , Zebrafish , Zinc/metabolism
5.
Curr Opin Cell Biol ; 67: 79-85, 2020 12.
Article En | MEDLINE | ID: mdl-32950922

Vertebrates sense a large variety of sensory stimuli that ranges from temperature, volatile and nonvolatile chemicals, touch, pain, light, sound and gravity. To achieve this, they use specialized cells present in sensory organs and cranial ganglia. Much of our understanding of the transcription factors and mechanisms responsible for sensory cell specification comes from cell-lineage tracing and genetic experiments in different species, but recent advances in single-cell transcriptomics, high-resolution imaging and systems biology approaches have allowed to study these processes in an unprecedented resolution. Here I will point to the transcription factor programs driving cell diversity in the different sensory organs of vertebrates to then discuss in vivo data of how cell specification is coupled with tissue morphogenesis.


Cell Differentiation , Skull/cytology , Vertebrates/physiology , Animals , Cell Lineage , Cellular Reprogramming , Humans , Morphogenesis
6.
Cell Rep ; 32(2): 107903, 2020 07 14.
Article En | MEDLINE | ID: mdl-32668260

In many organs, stem cell function depends on communication with their niche partners. Cranial sensory neurons develop in close proximity to blood vessels; however, whether vasculature is an integral component of their niches is yet unknown. Here, two separate roles for vasculature in cranial sensory neurogenesis in zebrafish are uncovered. The first involves precise spatiotemporal endothelial-neuroblast cytoneme contacts and Dll4-Notch signaling to restrain neuroblast proliferation. The second, instead, requires blood flow to trigger a transcriptional response that modifies neuroblast metabolic status and induces sensory neuron differentiation. In contrast, no role of sensory neurogenesis in vascular development is found, suggesting unidirectional signaling from vasculature to sensory neuroblasts. Altogether, we demonstrate that the cranial vasculature constitutes a niche component of the sensory ganglia that regulates the pace of their growth and differentiation dynamics.


Blood Circulation/physiology , Blood Vessels/cytology , Cell Cycle , Cell Differentiation , Sensory Receptor Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Circulation/drug effects , Body Patterning/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Count , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Oxygen/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Receptors, Notch/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction/drug effects , Skull/blood supply , Thiazolidines/pharmacology , Transcription, Genetic/drug effects , Vestibulocochlear Nerve/cytology , Vestibulocochlear Nerve/metabolism , Zebrafish , Zebrafish Proteins/metabolism
7.
J Anat ; 232(3): 431-439, 2018 03.
Article En | MEDLINE | ID: mdl-29235648

There is growing evidence of a direct influence of vasculature on the development of neurons in the brain. The development of the cranial vasculature has been well described in zebrafish but its anatomical relationship with the adjacent developing sensory ganglia has not been addressed. Here, by 3D imaging of fluorescently labelled blood vessels and sensory ganglia, we describe for the first time the spatial organization of the cranial vasculature in relation to the cranial ganglia during zebrafish development. We show that from 24 h post-fertilization (hpf) onwards, the statoacoustic ganglion (SAG) develops in direct contact with two main blood vessels, the primordial hindbrain channel and the lateral dorsal aortae (LDA). At 48 hpf, the LDA is displaced medially, losing direct contact with the SAG. The relationship of the other cranial ganglia with the vasculature is evident for the medial lateral line ganglion and for the vagal ganglia that grow along the primary head sinus (PHS). We also observed that the innervation of the anterior macula runs over the PHS vessel. Our spatiotemporal anatomical map of the cranial ganglia and the head vasculature indicates physical interactions between both systems and suggests a possible functional interaction during development.


Blood Vessels/embryology , Brain/blood supply , Brain/embryology , Cranial Nerves/blood supply , Zebrafish/embryology , Animals , Cranial Nerves/embryology , Ganglia/blood supply , Ganglia/embryology
8.
Elife ; 62017 05 24.
Article En | MEDLINE | ID: mdl-28537554

Neural patterning involves regionalised cell specification. Recent studies indicate that cell dynamics play instrumental roles in neural pattern refinement and progression, but the impact of cell behaviour and morphogenesis on neural specification is not understood. Here we combine 4D analysis of cell behaviours with dynamic quantification of proneural expression to uncover the construction of the zebrafish otic neurogenic domain. We identify pioneer cells expressing neurog1 outside the otic epithelium that migrate and ingress into the epithelialising placode to become the first otic neuronal progenitors. Subsequently, neighbouring cells express neurog1 inside the placode, and apical symmetric divisions amplify the specified pool. Interestingly, pioneer cells delaminate shortly after ingression. Ablation experiments reveal that pioneer cells promote neurog1 expression in other otic cells. Finally, ingression relies on the epithelialisation timing controlled by FGF activity. We propose a novel view for otic neurogenesis integrating cell dynamics whereby ingression of pioneer cells instructs neuronal specification.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Ear/embryology , Epithelium/embryology , Nerve Tissue Proteins/metabolism , Nervous System/embryology , Neuroepithelial Cells/physiology , Neurogenesis , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Body Patterning , Cell Movement
9.
Semin Cell Dev Biol ; 65: 47-59, 2017 05.
Article En | MEDLINE | ID: mdl-27686400

The vertebrate inner ear is a precision sensory organ, acting as both a microphone to receive sound and an accelerometer to detect gravity and motion. It consists of a series of interlinked, fluid-filled chambers containing patches of sensory epithelia, each with a specialised function. The ear contains many different differentiated cell types with distinct morphologies, from the flask-shaped hair cells found in thickened sensory epithelium, to the thin squamous cells that contribute to non-sensory structures, such as the semicircular canal ducts. Nearly all cell types of the inner ear, including the afferent neurons that innervate it, are derived from the otic placode, a region of cranial ectoderm that develops adjacent to the embryonic hindbrain. As the ear develops, the otic epithelia grow, fold, fuse and rearrange to form the complex three-dimensional shape of the membranous labyrinth. Much of our current understanding of the processes of inner ear morphogenesis comes from genetic and pharmacological manipulations of the developing ear in mouse, chicken and zebrafish embryos. These traditional approaches are now being supplemented with exciting new techniques-including force measurements and light-sheet microscopy-that are helping to elucidate the mechanisms that generate this intricate organ system.


Cell Lineage/genetics , Ectoderm/cytology , Epithelial Cells/cytology , Hair Cells, Auditory/cytology , Labyrinth Supporting Cells/cytology , Organogenesis/genetics , Animals , Cell Differentiation , Cell Movement , Chick Embryo , Ectoderm/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Labyrinth Supporting Cells/metabolism , Mice , Species Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish
10.
J Neurosci ; 35(47): 15752-66, 2015 Nov 25.
Article En | MEDLINE | ID: mdl-26609166

During development, otic sensory progenitors give rise to hair cells and supporting cells. In mammalian adults, differentiated and quiescent sensory cells are unable to generate new hair cells when these are lost due to various insults, leading to irreversible hearing loss. Retinoic acid (RA) has strong regenerative capacity in several organs, but its role in hair cell regeneration is unknown. Here, we use genetic and pharmacological inhibition to show that the RA pathway is required for hair cell regeneration in zebrafish. When regeneration is induced by laser ablation in the inner ear or by neomycin treatment in the lateral line, we observe rapid activation of several components of the RA pathway, with dynamics that position RA signaling upstream of other signaling pathways. We demonstrate that blockade of the RA pathway impairs cell proliferation of supporting cells in the inner ear and lateral line. Moreover, in neuromast, RA pathway regulates the transcription of p27(kip) and sox2 in supporting cells but not fgf3. Finally, genetic cell-lineage tracing using Kaede photoconversion demonstrates that de novo hair cells derive from FGF-active supporting cells. Our findings reveal that RA has a pivotal role in zebrafish hair cell regeneration by inducing supporting cell proliferation, and shed light on the underlying transcriptional mechanisms involved. This signaling pathway might be a promising approach for hearing recovery. SIGNIFICANCE STATEMENT: Hair cells are the specialized mechanosensory cells of the inner ear that capture auditory and balance sensory input. Hair cells die after acoustic trauma, ototoxic drugs or aging diseases, leading to progressive hearing loss. Mammals, in contrast to zebrafish, lack the ability to regenerate hair cells. Here, we find that retinoic acid (RA) pathway is required for hair cell regeneration in vivo in the zebrafish inner ear and lateral line. RA pathway is activated very early upon hair cell loss, promotes cell proliferation of progenitor cells, and regulates two key genes, p27(kip) and sox2. Our results position RA as an essential signal for hair cell regeneration with relevance in future regenerative strategies in mammals.


Cyclin-Dependent Kinase Inhibitor p27/metabolism , Hair Cells, Auditory/metabolism , Nerve Regeneration/physiology , SOX Transcription Factors/metabolism , Signal Transduction/physiology , Tretinoin/physiology , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cyclin-Dependent Kinase Inhibitor p27/antagonists & inhibitors , Female , Male , SOX Transcription Factors/antagonists & inhibitors , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
12.
Nat Commun ; 6: 7355, 2015 Jun 16.
Article En | MEDLINE | ID: mdl-26077034

Many organ functions rely on epithelial cavities with particular shapes. Morphogenetic anomalies in these cavities lead to kidney, brain or inner ear diseases. Despite their relevance, the mechanisms regulating lumen dimensions are poorly understood. Here, we perform live imaging of zebrafish inner ear development and quantitatively analyse the dynamics of lumen growth in 3D. Using genetic, chemical and mechanical interferences, we identify two new morphogenetic mechanisms underlying anisotropic lumen growth. The first mechanism involves thinning of the epithelium as the cells change their shape and lose fluids in concert with expansion of the cavity, suggesting an intra-organ fluid redistribution process. In the second mechanism, revealed by laser microsurgery experiments, mitotic rounding cells apicobasally contract the epithelium and mechanically contribute to expansion of the lumen. Since these mechanisms are axis specific, they not only regulate lumen growth but also the shape of the cavity.


Cell Shape , Ear, Inner/embryology , Epithelial Cells/cytology , Mitosis , Animals , Ear, Inner/cytology , Embryo, Nonmammalian , Imaging, Three-Dimensional , Intracellular Fluid/metabolism , Organogenesis , Zebrafish
13.
Mol Cell ; 58(5): 755-66, 2015 Jun 04.
Article En | MEDLINE | ID: mdl-25959397

Protein function is often regulated and controlled by posttranslational modifications, such as oxidation. Although oxidation has been mainly considered to be uncontrolled and nonenzymatic, many enzymatic oxidations occur on enzyme-selected lysine residues; for instance, LOXL2 oxidizes lysines by converting the ε-amino groups into aldehyde groups. Using an unbiased proteomic approach, we have identified methylated TAF10, a member of the TFIID complex, as a LOXL2 substrate. LOXL2 oxidation of TAF10 induces its release from its promoters, leading to a block in TFIID-dependent gene transcription. In embryonic stem cells, this results in the inactivation of the pluripotency genes and loss of the pluripotent capacity. During zebrafish development, the absence of LOXL2 resulted in the aberrant overexpression of the neural progenitor gene Sox2 and impaired neural differentiation. Thus, lysine oxidation of the transcription factor TAF10 is a controlled protein modification and demonstrates a role for protein oxidation in regulating pluripotency genes.


Amino Acid Oxidoreductases/physiology , Cell Differentiation , Neural Stem Cells/physiology , Protein Processing, Post-Translational , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/physiology , Animals , Epigenesis, Genetic , HEK293 Cells , Humans , Methylation , Oxidation-Reduction , Transcription Factor TFIID/metabolism , Zebrafish
14.
PLoS One ; 9(10): e109860, 2014.
Article En | MEDLINE | ID: mdl-25299450

The generation of sensory neurons and hair cells of the inner ear is under tight control. Different members of the Hairy and Enhancer of Split genes (HES) are expressed in the inner ear, their full array of functions still not being disclosed. We have previously shown that zebrafish her9 acts as a patterning gene to restrict otic neurogenesis to an anterior domain. Here, we disclose the role of another her gene, her4, a zebrafish ortholog of Hes5 that is expressed in the neurogenic and sensory domains of the inner ear. The expression of her4 is highly dynamic and spatiotemporally regulated. We demonstrate by loss of function experiments that in the neurogenic domain her4 expression is under the regulation of neurogenin1 (neurog1) and the Notch pathway. Moreover, her4 participates in lateral inhibition during otic neurogenesis since her4 knockdown results in overproduction of the number of neurog1 and deltaB-positive otic neurons. In contrast, during sensorigenesis her4 is initially Notch-independent and induced by atoh1b in a broad prosensory domain. At later stages her4 expression becomes Notch-dependent in the future sensory domains but loss of her4 does not result in hair cell overproduction, suggesting that there other her genes can compensate its function.


Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Ear, Inner/growth & development , Embryonic Development , Neurogenesis , Zebrafish Proteins/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Ear, Inner/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Hair Cells, Auditory, Inner/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, Notch/biosynthesis , Receptors, Notch/genetics , Sensory Receptor Cells/metabolism , Signal Transduction/genetics , Zebrafish , Zebrafish Proteins/genetics
15.
Dev Biol ; 389(1): 50-67, 2014 May 01.
Article En | MEDLINE | ID: mdl-24508480

For both the intricate morphogenetic layout of the sensory cells in the ear and the elegantly radial arrangement of the sensory neurons in the nose, numerous signaling molecules and genetic determinants are required in concert to generate these specialized neuronal populations that help connect us to our environment. In this review, we outline many of the proteins and pathways that play essential roles in the differentiation of otic and olfactory neurons and their integration into their non-neuronal support structures. In both cases, well-known signaling pathways together with region-specific factors transform thickened ectodermal placodes into complex sense organs containing numerous, diverse neuronal subtypes. Olfactory and otic placodes, in combination with migratory neural crest stem cells, generate highly specialized subtypes of neuronal cells that sense sound, position and movement in space, odors and pheromones throughout our lives.


Ear, Inner/embryology , Ectoderm/embryology , Neurogenesis/physiology , Olfactory Pathways/embryology , Sense Organs/embryology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Ear, Inner/cytology , Ear, Inner/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Gene Expression Regulation, Developmental , Humans , Neurogenesis/genetics , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Sense Organs/cytology , Sense Organs/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
16.
J Cell Sci ; 126(Pt 1): 53-9, 2013 Jan 01.
Article En | MEDLINE | ID: mdl-23178937

In a genome-wide expression profile search for genes required for Drosophila R7 photoreceptor development we found ß amyloid protein precursor-like (Appl), the ortholog of human APP, which is a key factor in the pathogenesis of Alzheimer's disease. We analyzed Appl expression in the eye imaginal disc and found that is highly accumulated in R7 photoreceptor cells. The R7 photoreceptor is responsible for UV light detection. To explore the link between high expression of Appl and R7 function, we have analyzed Appl null mutants and found reduced preference for UV light, probably because of mistargeted R7 axons. Moreover, axon mistargeting and inappropriate light discrimination are enhanced in combination with neurotactin mutants. R7 differentiation is triggered by the inductive interaction between R8 and R7 precursors, which results in a burst of Ras1/MAPK, activated by the tyrosine kinase receptor Sevenless. Therefore, we examined whether Ras1/MAPK is responsible for the high Appl expression. Inhibition of Ras1 signaling leads to reduced Appl expression, whereas constitutive activation drives ectopic Appl expression. We show that Appl is directly regulated by the Ras/MAPK pathway through a mechanism mediated by PntP2, an ETS transcription factor that specifically binds ETS sites in the Appl regulatory region. We also found that zebrafish appb expression increased after ectopic fgfr activation in the neural tube of zebrafish embryos, suggesting a conserved regulatory mechanism.


Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , ras Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , ras Proteins/genetics
17.
Development ; 138(3): 397-408, 2011 Feb.
Article En | MEDLINE | ID: mdl-21205785

Proper spatial control of neurogenesis in the inner ear ensures the precise innervation of mechanotransducing cells and the propagation of auditory and equilibrium stimuli to the brain. Members of the Hairy and enhancer of split (Hes) gene family regulate neurogenesis by inhibiting neuronal differentiation and maintaining neural stem cell pools in non-neurogenic zones. Remarkably, their role in the spatial control of neurogenesis in the ear is unknown. In this study, we identify her9, a zebrafish ortholog of Hes1, as a key gene in regulating otic neurogenesis through the definition of the posterolateral non-neurogenic field. First, her9 emerges as a novel otic patterning gene that represses proneural function and regulates the extent of the neurogenic domain. Second, we place Her9 downstream of Tbx1, linking these two families of transcription factors for the first time in the inner ear and suggesting that the reported role of Tbx1 in repressing neurogenesis is in part mediated by the bHLH transcriptional repressor Her9. Third, we have identified retinoic acid (RA) signaling as the upstream patterning signal of otic posterolateral genes such as tbx1 and her9. Finally, we show that at the level of the cranial otic field, opposing RA and Hedgehog signaling position the boundary between the neurogenic and non-neurogenic compartments. These findings permit modeling of the complex genetic cascade that underlies neural patterning of the otic vesicle.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , T-Box Domain Proteins/metabolism , Tretinoin/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation/drug effects , Ear, Inner , Embryo, Nonmammalian/drug effects , Immunohistochemistry , In Situ Hybridization , T-Box Domain Proteins/genetics , Tretinoin/pharmacology , Zebrafish , Zebrafish Proteins/genetics
18.
Hum Genet ; 128(4): 411-9, 2010 Oct.
Article En | MEDLINE | ID: mdl-20668882

POU3F4 encodes a POU-domain transcription factor required for inner ear development. Defects in POU3F4 function are associated with X-linked deafness type 3 (DFN3). Multiple deletions affecting up to ~900-kb upstream of POU3F4 are found in DFN3 patients, suggesting the presence of essential POU3F4 enhancers in this region. Recently, an inner ear enhancer was reported that is absent in most DFN3 patients with upstream deletions. However, two indications suggest that additional enhancers in the POU3F4 upstream region are required for POU3F4 function during inner ear development. First, there is at least one DFN3 deletion that does not eliminate the reported enhancer. Second, the expression pattern driven by this enhancer does not fully recapitulate Pou3f4 expression in the inner ear. Here, we screened a 1-Mb region upstream of the POU3F4 gene for additional cis-regulatory elements and searched for novel DFN3 mutations in the identified POU3F4 enhancers. We found several novel enhancers for otic vesicle expression. Some of these also drive expression in kidney, pancreas and brain, tissues that are known to express Pou3f4. In addition, we report a new and smallest deletion identified so far in a DFN3 family which eliminates 3.9 kb, comprising almost exclusively the previous reported inner ear enhancer. We suggest that multiple enhancers control the expression of Pou3f4 in the inner ear and these may contribute to the phenotype observed in DFN3 patients. In addition, the novel deletion demonstrates that the previous reported enhancer, although not sufficient, is essential for POU3F4 function during inner ear development.


5' Flanking Region/genetics , Ear, Inner/metabolism , Enhancer Elements, Genetic/genetics , Hearing/genetics , POU Domain Factors/genetics , Animals , Base Sequence , DNA Mutational Analysis , Ear, Inner/growth & development , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Family Health , Female , Gene Deletion , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hearing Loss/genetics , Humans , In Situ Hybridization , Male , Microscopy, Fluorescence , Pedigree , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Xenopus/embryology , Xenopus/genetics
19.
PLoS One ; 5(12): e15907, 2010 Dec 31.
Article En | MEDLINE | ID: mdl-21209840

POU3F4 is a member of the POU-homedomain transcription factor family with a prominent role in inner ear development. Mutations in the human POU3F4 coding unit leads to X-linked deafness type 3 (DFN3), characterized by conductive hearing loss and progressive sensorineural deafness. Microdeletions found 1 Mb 5' upstream of the coding region also displayed the same phenotype, suggesting that cis-regulatory elements might be present in that region. Indeed, we and others have recently identified several enhancers at the 1 Mb 5' upstream interval of the pou3f4 locus. Here we characterize the spatio-temporal patterns of these regulatory elements in zebrafish transgenic lines. We show that the most distal enhancer (HCNR 81675) is activated earlier and drives GFP reporter expression initially to a broad ear domain to progressively restrict to the sensory patches. The proximal enhancer (HCNR 82478) is switched later during development and promotes expression, among in other tissues, in sensory patches from its onset. The third enhancer (HCNR 81728) is also active at later stages in the otic mesenchyme and in the otic epithelium. We also characterize the signaling pathways regulating these enhancers. While HCNR 81675 is regulated by very early signals of retinoic acid, HCNR 82478 is regulated by Fgf activity at a later stage and the HCNR 81728 enhancer is under the control of Hh signaling. Finally, we show that Sox2 and Pax2 transcription factors are bound to HCNR 81675 genomic region during otic development and specific mutations to these transcription factor binding sites abrogates HCNR 81675 enhancer activity. Altogether, our results suggest that pou3f4 expression in inner ear might be under the control of distinct regulatory elements that fine-tune the spatio-temporal activity of this gene and provides novel data on the signaling mechanisms controlling pou3f4 function.


POU Domain Factors/genetics , Animals , Binding Sites , Ear/embryology , Enhancer Elements, Genetic , Gene Deletion , Green Fluorescent Proteins/metabolism , Humans , Mice , Models, Genetic , Mutation , Time Factors , Transcription Factors/metabolism , Xenopus , Zebrafish
20.
Int J Dev Biol ; 53(8-10): 1503-13, 2009.
Article En | MEDLINE | ID: mdl-19247974

The inner ear is a complex structure responsible for the senses of audition and balance in vertebrates. The ear is organised into different sense organs that are specialised to detect specific stimuli such as sound and linear or angular accelerations. The elementary sensory unit of the ear consists of hair cells, supporting cells, neurons and Schwann cells. Hair cells are the mechano-electrical transducing elements, and otic neurons convey information coded in electrical impulses to the brain. With the exception of the Schwann cells, all cellular elements of the inner ear derive from the otic placode. This is an ectodermal thickening that is specified in the head ectoderm adjacent to the caudal hindbrain. The complex organisation of the ear requires precise coupling of regional specification and cell fate decisions during development, i.e. specificity in defining particular spatial domains containing particular cell types. Those decisions are taken early in development and are the subject of this article. We review here recent work on: i) early patterning of the otic placode, ii) the role of neural tube signals in the patterning of the otic vesicle, and iii) the genes underlying cell fate determination of neurons and sensory hair cells.


Body Patterning/genetics , Ear, Inner/embryology , Ear, Inner/metabolism , Gene Expression Regulation, Developmental , Animals , Cell Differentiation/genetics , Chickens , Ear, Inner/cytology , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Mice , Models, Biological , Rhombencephalon/embryology , Rhombencephalon/metabolism
...