Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Pharm ; 574: 118734, 2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31705970

ABSTRACT

Achieving drug delivery at the ocular level encounters many challenges and obstacles. In situ gelling delivery systems are now widely used for topical ocular administration and recognized as a promising strategy to improve the treatment of a wide range of ocular diseases. The present work describes the formulation and evaluation of a mucoadhesive and ion-activated in situ gelling delivery system based on gellan gum and hydroxyethylcellulose for the delivery of phenylephrine and tropicamide. First, physico-chemical characteristics were assessed to ensure suitable properties regarding ocular administration. Then, rheological properties such as viscosity and gelation capacity were determined. Gelation capacity of the formulations and the effect of hydroxyethylcellulose on viscosity were demonstrated. A new rheological method was developed to assess the gel resistance under simulated eye blinking. Afterward, mucoadhesion was evaluated using tensile strength test and rheological synergism method in both rotational and oscillatory mode allowing mucoadhesive properties of hydroxyethylcellulose to be point out. Finally, residence time on the ocular surface was investigated in vivo, using cyanine 5.5 dye as a fluorescent marker entrapped in the in situ gelling delivery systems. Residence performance was studied by non-invasive optical imaging on vigilant rabbits, allowing eye blinking and nasolacrimal drainage to occur physiologically. Fluorescence intensity profiles pointed out a prolonged residence time on the ocular surface region for the developed formulations compared to conventional eye drops, suggesting in vitro / in vivo correlations between rheological properties and in vivo residence performances.


Subject(s)
Cellulose/analogs & derivatives , Cornea/drug effects , Gels/chemistry , Gels/pharmacology , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacology , Polysaccharides, Bacterial/chemistry , Administration, Ophthalmic , Animals , Biological Availability , Cellulose/chemistry , Drug Delivery Systems/methods , Drug Liberation , Excipients/chemistry , Male , Rabbits , Rheology/methods , Viscosity
2.
Dev Cell ; 42(5): 527-541.e4, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28867488

ABSTRACT

Skeletal muscle contraction is mediated by myofibrils, complex multi-molecular scaffolds structured into repeated units, the sarcomeres. Myofibril structure and function have been extensively studied, but the molecular processes regulating its formation within the differentiating muscle cell remain largely unknown. Here we show in zebrafish that genetic interference with the Quaking RNA-binding proteins disrupts the initial steps of myofibril assembly without affecting early muscle differentiation. Using RNA sequencing, we demonstrate that Quaking is required for accumulation of the muscle-specific tropomyosin-3 transcript, tpm3.12. Further functional analyses reveal that Tpm3.12 mediates Quaking control of myofibril formation. Moreover, we identified a Quaking-binding site in the 3' UTR of tpm3.12 transcript, which is required in vivo for tpm3.12 accumulation and myofibril formation. Our work uncovers a Quaking/Tpm3 pathway controlling de novo myofibril assembly. This unexpected developmental role for Tpm3 could be at the origin of muscle defects observed in human congenital myopathies associated with tpm3 mutation.


Subject(s)
Myofibrils/metabolism , RNA-Binding Proteins/metabolism , Tropomyosin/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , 3' Untranslated Regions/genetics , Animals , Binding Sites , Cell Differentiation/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/genetics , Myosins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcomeres/metabolism , Somites/embryology , Somites/metabolism , Zebrafish/embryology , Zebrafish/genetics
3.
Dev Neurobiol ; 75(9): 961-83, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25556989

ABSTRACT

MYO7A is an unconventional myosin involved in the structural organization of hair bundles at the apex of sensory hair cells (SHCs) where it serves mechanotransduction in the process of hearing and balance. Mutations of MYO7A are responsible for abnormal shaping of hair bundles, resulting in human deafness and murine deafness/circling behavior. Myo7aa, expressed in SHCs of the inner ear and lateral line of zebrafish, causes circling behavior and abnormal hair cell function when deficient in mariner mutant. This work identifies a new hair cell-specific enhancer, highly conserved between species, located within Intron 2-3 of zebrafish myosin 7a (myo7aa) gene. This enhancer is contained within a 761-bp DNA fragment that encompasses a newly identified Exon of myo7aa and whose activity does not depend on orientation. Compensation of mariner mutation by expression of mCherry-Myo7aa fusion protein under the control of this 761-bp DNA fragment results in recovery of balance, normal hair bundle shape and restored hair cell function. Two smaller adjacent fragments (344-bp and 431-bp), extracted from the 761-bp fragment, both show hair cell-specific enhancing activity, with apparently reduced intensity and coverage. These data should help understand the role of Myo7aa in sensory hair cell differentiation and function. They provide tools to decipher how myo7aa gene is expressed and regulated in SHCs by allowing the identification of potential transcription factors involved in this process. The discovered enhancer could represent a new target for the identification of deafness-causing mutations affecting human MYO7A.


Subject(s)
Enhancer Elements, Genetic , Exons , Hair Cells, Auditory/metabolism , Lateral Line System/cytology , Mechanoreceptors/metabolism , Myosins/genetics , Myosins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Introns , Lateral Line System/embryology , Lateral Line System/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutation , Postural Balance/physiology , RNA, Messenger/metabolism , Reflex, Startle/physiology , Sequence Homology , Zebrafish , Red Fluorescent Protein
4.
Front Behav Neurosci ; 7: 51, 2013.
Article in English | MEDLINE | ID: mdl-23720615

ABSTRACT

As soon as zebrafish larvae start eating, they exhibit a marked aversion for bitter and acidic substances, as revealed by a consumption assay, in which fluorescent Tetrahymena serve as a feeding basis, to which various stimuli can be added. Bitter and acidic substances elicited an increase in mRNA accumulation of the immediate-early response gene egr-1, as revealed by in situ hybridization. Conversely, chemostimulants that did not induce aversion did not induce egr-1 response. Maximum labeling was observed in cells located in the oropharyngeal cavity and on the gill rakers. Gustatory areas of the brain were also labeled. Interestingly, when bitter tastants were repeatedly associated with food reward, zebrafish juveniles learned to ingest food in the presence of the bitter compound. After habituation, the acquisition of acceptance for bitterness was accompanied by a loss of egr-1 labeling. Altogether, our data indicate that egr-1 participates specifically in food aversion. The existence of reward-coupled changes in taste sensitivity in humans suggests that our results are relevant to situations in humans.

5.
Development ; 140(5): 1090-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364327

ABSTRACT

Endoderm and mesoderm are both formed upon activation of Nodal signaling but how endoderm differentiates from mesoderm is still poorly explored. The sox-related gene casanova (sox32) acts downstream of the Nodal signal, is essential for endoderm development and requires the co-factor Pou2 (Pou5f1, Oct3, Oct4) in this process. Conversely, BMP signals have been shown to inhibit endoderm development by an as yet unexplained mechanism. In a search for Casanova regulators in zebrafish, we identified two of its binding partners as the transcription factors Pou2 and Vox, a member of the Vent group of proteins also involved in the patterning of the gastrula. In overexpression studies we show that vox and/or Vent group genes inhibit the capacity of Casanova to induce endoderm, even in the presence of its co-factor Pou2, and that Vox acts as a repressor in this process. We further show that vox, but not other members of the Vent group, is essential for defining the proper endodermal domain size at gastrulation. In this process, vox acts downstream of BMPs. Cell fate analysis further shows that Vox plays a key role downstream of BMP signals in regulating the capacity of Nodal to induce endoderm versus mesoderm by modulating the activity of the Casanova/Pou2 regulatory system.


Subject(s)
Endoderm/embryology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Octamer Transcription Factor-3/metabolism , Repressor Proteins/metabolism , Repressor Proteins/physiology , SOX Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Down-Regulation/genetics , Embryo, Nonmammalian , Endoderm/growth & development , Endoderm/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Nodal Signaling Ligands/genetics , Nodal Signaling Ligands/metabolism , Nodal Signaling Ligands/physiology , Octamer Transcription Factor-3/physiology , Protein Binding/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Repressor Proteins/chemistry , Repressor Proteins/genetics , SOX Transcription Factors/physiology , Sequence Deletion , Tissue Distribution , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 109(42): 16945-50, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027928

ABSTRACT

Collective cell migration is key to morphogenesis, wound healing, or cancer cell migration. However, its cellular bases are just starting to be unraveled. During vertebrate gastrulation, axial mesendoderm migrates in a group, the prechordal plate, from the embryonic organizer to the animal pole. How this collective migration is achieved remains unclear. Previous work has suggested that cells migrate as individuals, with collective movement resulting from the addition of similar individual cell behavior. Through extensive analyses of cell trajectories, morphologies, and polarization in zebrafish embryos, we reveal that all prechordal plate cells show the same behavior and rely on the same signaling pathway to migrate, as expected if they do so individually. However, by using cell transplants, we demonstrate that prechordal plate migration is a true collective process, as isolated cells do not migrate toward the animal pole. They are still polarized and motile but lose directionality. Directionality is restored upon contact with the endogenous prechordal plate. This contact dependent orientation relies on E-cadherin, Wnt-PCP signaling, and Rac1. Importantly, groups of cells also need contact with the endogenous plate to orient correctly, showing an instructive role of the plate in establishing directionality. Overall, our results lead to an original model of collective migration in which directional information is contained within the moving group rather than provided by extrinsic cues, and constantly maintained in cells by contacts with their neighbors. This self-organizing model could account for collective invasion of new territories, as observed in cancer strands, without requirement for any attractant in the colonized tissue.


Subject(s)
Cell Movement/physiology , Endoderm/physiology , Mesoderm/physiology , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Cadherins/metabolism , Cell Polarity/physiology , Endoderm/cytology , In Situ Hybridization , Mesoderm/cytology , Time-Lapse Imaging , Wnt Signaling Pathway/physiology , Zebrafish
7.
Development ; 138(16): 3473-84, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21791527

ABSTRACT

Taste buds, the taste sensory organs, are conserved in vertebrates and composed of distinct cell types, including taste receptor, basal/presynaptic and support cells. Here, we characterize zebrafish taste bud development and show that compromised Fgf signaling in the larva results in taste bud reduction and disorganization. We determine that Fgf activity is required within pharyngeal endoderm for formation of Calb2b(+) cells and reveal miR-200 and Delta-Notch signaling as key factors in this process. miR-200 knock down shows that miR-200 activity is required for taste bud formation and in particular for Calb2b(+) cell formation. Compromised delta activity in mib(-/-) dramatically reduces the number of Calb2b(+) cells and increases the number of 5HT(+) cells. Conversely, larvae with increased Notch activity and ascl1a(-/-) mutants are devoid of 5HT(+) cells, but have maintained and increased Calb2b(+) cells, respectively. These results show that Delta-Notch signaling is required for intact taste bud organ formation. Consistent with this, Notch activity restores Calb2b(+) cell formation in pharyngeal endoderm with compromised Fgf signaling, but fails to restore the formation of these cells after miR-200 knock down. Altogether, this study provides genetic evidence that supports a novel model where Fgf regulates Delta-Notch signaling, and subsequently miR-200 activity, in order to promote taste bud cell type differentiation.


Subject(s)
MicroRNAs/genetics , Signal Transduction , Taste Buds/embryology , Taste Buds/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibroblast Growth Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Taste Buds/growth & development , Transcription Factors , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
Development ; 138(9): 1783-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21447554

ABSTRACT

The development of the different muscles within the somite is a complex process that involves the Hedgehog (Hh) signaling pathway. To specify the proper number of muscle cells and organize them spatially and temporally, the Hh signaling pathway needs to be precisely regulated at different levels, but only a few factors external to the pathway have been described. Here, we report for the first time the role of the STAR family RNA-binding protein Quaking A (QkA) in somite muscle development. We show in zebrafish that the loss of QkA function affects fast muscle fiber maturation as well as Hh-induced muscle derivative specification and/or morphogenesis. Mosaic analysis reveals that fast fiber maturation depends on the activity of QkA in the environment of fast fiber progenitors. We further show that Hh signaling requires QkA activity for muscle development. By an in silico approach, we screened the 3'UTRs of known Hh signaling component mRNAs for the Quaking response element and found the transcription factor Gli2a, a known regulator of muscle fate development. Using destabilized GFP as a reporter, we show that the gli2a mRNA 3'UTR is a functional QkA target. Consistent with this notion, the loss of QkA function rescued slow muscle fibers in yot mutant embryos, which express a dominant-negative Gli2a isoform. Thus, our results reveal a new mechanism to ensure muscle cell fate diversity by fine-tuning of the Hh signaling pathway via RNA-binding proteins.


Subject(s)
Hedgehog Proteins/physiology , Muscle Development/genetics , RNA-Binding Proteins/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Chromosome Mapping , Embryo, Nonmammalian , Genes, Recessive , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Muscle Development/physiology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/physiology , Mutation/physiology , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
9.
Mol Cancer Res ; 7(3): 402-14, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19276186

ABSTRACT

Overexpression of the ERBB2 gene, linked to genomic and transcriptional amplifications, is a poor prognosis indicator in 25% to 30% of breast cancers. In contrast to some well-documented genomic amplifications, molecular mechanisms leading to ERBB2 transcriptional overexpression remain poorly characterized. Gene expression analyses of breast cancer have characterized distinct transcriptional signatures allowing a molecular classification of breast carcinoma. Coexpression of the ERBB2 and GATA4 genes was originally observed in tumors. Both genes are essential for cardiovascular development and GATA4 has been proposed to control the transcription of critical genes for the differentiation and the function of myocardium. We determined that ERBB2-targeted small interfering RNA repressed both ERBB2 and GATA4 genes, whereas GATA4-targeted small interfering RNA repressed GATA4 and activated ERBB2 transcription. Transfected GATA4-expressing construct repressed ERBB2 promoter. Phylogenetic foot printing revealed multiple putative GATA4 binding sites conserved in mammals within the ERBB2 promoter region. Chromatin immunoprecipitation showed that GATA4 binds specifically to several ERBB2 gene noncoding regions. Electrophoretic mobility shift assay revealed GATA4 binding to a well-conserved consensus motif. Site-directed mutagenesis confirmed the role of this new regulatory element for the activity of the ERBB2 gene enhancer. In agreement with a repressor role of GATA4 on ERBB2 gene expression balanced by ERBB2 activation of the GATA4 gene, a negative correlation between the relative levels of ERBB2 and GATA4 mRNA was observed in breast cancer cell lines and breast tumor samples. We propose that the negative feedback loop linking ERBB2 and GATA4 plays a role in the transcriptional dysregulation of ERBB2 gene expression in breast cancer.


Subject(s)
Breast Neoplasms/genetics , GATA4 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/genetics , Binding Sites , Breast Neoplasms/metabolism , Cell Line, Tumor , Conserved Sequence , Feedback, Physiological , GATA4 Transcription Factor/biosynthesis , Humans , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, ErbB-2/biosynthesis , Transcriptional Activation , Transfection
10.
Curr Biol ; 18(4): 276-81, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18291651

ABSTRACT

During gastrulation, dramatic movements rearrange cells into three germ layers expanded over the entire embryo [1-3]. In fish, both endoderm and mesoderm are specified as a belt at the embryo margin. Mesodermal layer expansion is achieved through the combination of two directed migrations. The outer ring of precursors moves toward the vegetal pole and continuously seeds mesodermal cells inside the embryo, which then reverse their movement in the direction of the animal pole [3-6]. Unlike mesoderm, endodermal cells internalize at once and must therefore adopt a different strategy to expand over the embryo [7, 8]. With live imaging of YFP-expressing zebrafish endodermal cells, we demonstrate that in contrast to mesoderm, internalized endodermal cells display a nonoriented/noncoordinated movement fit by a random walk that rapidly disperses them over the yolk surface. Transplantation experiments reveal that this behaviour is largely cell autonomous, induced by TGF-beta/Nodal, and dependent on the downstream effector Casanova. At midgastrulation, endodermal cells switch to a convergence movement. We demonstrate that this switch is triggered by environmental cues. These results uncover random walk as a novel Nodal-induced gastrulation movement and as an efficient strategy to transform a localized cell group into a layer expanded over the embryo.


Subject(s)
Cell Movement/physiology , Endoderm/cytology , Gastrulation/physiology , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/physiology , Embryonic Induction/physiology , Endoderm/physiology , High Mobility Group Proteins/metabolism , Nodal Protein , SOX Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Zebrafish/physiology , Zebrafish Proteins/metabolism
11.
PLoS One ; 3(1): e1434, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18197245

ABSTRACT

The EGF-CFC factor Oep/Cripto1/Frl1 has been implicated in embryogenesis and several human cancers. During vertebrate development, Oep/Cripto1/Frl1 has been shown to act as an essential coreceptor in the TGFbeta/Nodal pathway, which is crucial for germ layer formation. Although studies in cell cultures suggest that Oep/Cripto1/Frl1 is also implicated in other pathways, in vivo it is solely regarded as a Nodal coreceptor. We have found that Rasl11b, a small GTPase belonging to a Ras subfamily of putative tumor suppressor genes, modulates Oep function in zebrafish independently of the Nodal pathway. rasl11b down regulation partially rescues endodermal and prechordal plate defects of zygotic oep(-/-) mutants (Zoep). Rasl11b inhibitory action was only observed in oep-deficient backgrounds, suggesting that normal oep expression prevents Rasl11b function. Surprisingly, rasl11b down regulation does not rescue mesendodermal defects in other Nodal pathway mutants, nor does it influence the phosphorylation state of the downstream effector Smad2. Thus, Rasl11b modifies the effect of Oep on mesendoderm development independently of the main known Oep output: the Nodal signaling pathway. This data suggests a new branch of Oep signaling that has implications for germ layer development, as well as for studies of Oep/Frl1/Cripto1 dysfunction, such as that found in tumors.


Subject(s)
Homeodomain Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Base Sequence , DNA Primers , Mutagenesis , Phenotype , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Smad2 Protein/metabolism , Zebrafish
12.
PLoS Biol ; 5(10): e277, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17941721

ABSTRACT

Sleep is a fundamental biological process conserved across the animal kingdom. The study of how sleep regulatory networks are conserved is needed to better understand sleep across evolution. We present a detailed description of a sleep state in adult zebrafish characterized by reversible periods of immobility, increased arousal threshold, and place preference. Rest deprivation using gentle electrical stimulation is followed by a sleep rebound, indicating homeostatic regulation. In contrast to mammals and similarly to birds, light suppresses sleep in zebrafish, with no evidence for a sleep rebound. We also identify a null mutation in the sole receptor for the wake-promoting neuropeptide hypocretin (orexin) in zebrafish. Fish lacking this receptor demonstrate short and fragmented sleep in the dark, in striking contrast to the excessive sleepiness and cataplexy of narcolepsy in mammals. Consistent with this observation, we find that the hypocretin receptor does not colocalize with known major wake-promoting monoaminergic and cholinergic cell groups in the zebrafish. Instead, it colocalizes with large populations of GABAergic neurons, including a subpopulation of Adra2a-positive GABAergic cells in the anterior hypothalamic area, neurons that could assume a sleep modulatory role. Our study validates the use of zebrafish for the study of sleep and indicates molecular diversity in sleep regulatory networks across vertebrates.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Sleep Initiation and Maintenance Disorders/metabolism , Sleep/physiology , Zebrafish Proteins/metabolism , Zebrafish/physiology , Amino Acid Sequence , Animals , Arousal/physiology , Behavior, Animal/physiology , Biogenic Monoamines/metabolism , Brain/anatomy & histology , Brain/metabolism , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Light , Molecular Sequence Data , Neuropeptides/metabolism , Orexin Receptors , Orexins , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , Sequence Alignment , Sleep Deprivation , Zebrafish/anatomy & histology , Zebrafish/embryology , Zebrafish Proteins/genetics
13.
Genome Biol ; 8(8): R173, 2007.
Article in English | MEDLINE | ID: mdl-17711588

ABSTRACT

BACKGROUND: MicroRNA (miRNA) encoding genes are abundant in vertebrate genomes but very few have been studied in any detail. Bioinformatic tools allow prediction of miRNA targets and this information coupled with knowledge of miRNA expression profiles facilitates formulation of hypotheses of miRNA function. Although the central nervous system (CNS) is a prominent site of miRNA expression, virtually nothing is known about the spatial and temporal expression profiles of miRNAs in the brain. To provide an overview of the breadth of miRNA expression in the CNS, we performed a comprehensive analysis of the neuroanatomical expression profiles of 38 abundant conserved miRNAs in developing and adult zebrafish brain. RESULTS: Our results show miRNAs have a wide variety of different expression profiles in neural cells, including: expression in neuronal precursors and stem cells (for example, miR-92b); expression associated with transition from proliferation to differentiation (for example, miR-124); constitutive expression in mature neurons (miR-124 again); expression in both proliferative cells and their differentiated progeny (for example, miR-9); regionally restricted expression (for example, miR-222 in telencephalon); and cell-type specific expression (for example, miR-218a in motor neurons). CONCLUSION: The data we present facilitate prediction of likely modes of miRNA function in the CNS and many miRNA expression profiles are consistent with the mutual exclusion mode of function in which there is spatial or temporal exclusion of miRNAs and their targets. However, some miRNAs, such as those with cell-type specific expression, are more likely to be co-expressed with their targets. Our data provide an important resource for future functional studies of miRNAs in the CNS.


Subject(s)
Brain/growth & development , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Organogenesis/genetics , Zebrafish/growth & development , Animals , Brain/cytology , Cell Differentiation/genetics , Gene Expression Profiling , Larva/chemistry , Larva/cytology , Larva/genetics , Larva/growth & development , MicroRNAs/analysis , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Zebrafish/genetics
14.
Pharmacogenet Genomics ; 17(4): 237-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17496723

ABSTRACT

OBJECTIVES: The zebrafish is an ideally suited vertebrate animal model for large-scale genetic screens and is emerging as a model organism in pharmacological and behavioral research. We investigated the effects of sedative hypnotics commonly used in humans on zebrafish locomotor activity and identified the corresponding genomic and receptor binding targets. METHODS: We studied radioreceptor binding and behavioral responses to compounds with known sedative hypnotic properties representing multiple pharmacological classes. These included GABAergic hypnotics such as benzodiazepines, barbiturates, and baclofen; alpha-2 adrenergic agonists; and histaminergic H1 antagonists. An automated system was used to quantify behavioral effects. Zebrafish homologs of histamine receptor H1, gamma-amino-n-butyric acid type A (alpha-subunit), and gamma-amino-n-butyric acid type B (1 and 2) receptor genes were identified through translating queries of the zebrafish Zv4 database with human receptor protein sequences. A pilot screen of 154 N-ethyl-N-nitroso-urea-mutagenized F2 families was conducted with pentobarbital, flurazepam and mepyramine. RESULTS: Radioreceptor binding studies revealed high affinity binding sites for known gamma-amino-n-butyric acid type A, gamma-amino-n-butyric acid type B, and histaminergic ligands. Drug immersion of 5-7-day-old larvae reduced mobility and, in some cases, produced a complete state of unresponsive immobility similar to anesthesia. These effects were dose-dependent and rapidly reversible in water. As established in mammals, (R)-baclofen was more active behaviorally and had higher affinity in binding studies when compared with (S)-baclofen. In this model, (S)-baclofen only partially reduced activity at high dose and blocked (R)-baclofen behavioral hypnotic effects. Genomic sequences with high similarity to the corresponding pharmacological targets were identified, but no mutants were found in the pilot screen. CONCLUSIONS: These results demonstrate conservation of gene, protein and function for many established sedative hypnotic pathways. The results indicate feasibility of conducting large-scale pharmacogenomic screens to isolate novel proteins modulating susceptibility to hypnotic compounds in a vertebrate system.


Subject(s)
Hypnotics and Sedatives/pharmacology , Zebrafish/genetics , Zebrafish/physiology , Amino Acid Sequence , Animals , Baclofen/chemistry , Baclofen/pharmacology , Behavior, Animal/drug effects , Binding Sites/genetics , Conserved Sequence , Humans , Hypnotics and Sedatives/chemistry , Larva/drug effects , Larva/metabolism , Larva/physiology , Molecular Sequence Data , Motor Activity/drug effects , Motor Activity/genetics , Pharmacogenetics , Phylogeny , Radioligand Assay , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-B/drug effects , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Receptors, Histamine H1/drug effects , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Sequence Homology, Amino Acid , Zebrafish/metabolism
15.
Mol Cell Neurosci ; 33(2): 180-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16949838

ABSTRACT

In this report, we present a study of regeneration of the lateral line, a collection of mechano-sensory organ, in the adult zebrafish caudal fin. As all neuromasts are innervated by axon fibers, neuronal regeneration is a key issue in the regenerating process. We first show that support cells from the last neuromast adjacent to the amputation plane divide and migrate to colonize the blastema in order to reform the missing part of the lateral line. We then show that nerve re-growth takes place later than neuromast progenitor cell migration. We also provide evidence that new growth cones form at the amputation plane and subsequently follow the migrating placode-like structure to re-innervate regenerated neuromasts as they differentiate. Altogether, our observations indicate that caudal lateral line regeneration is not a mere recapitulation of the ontogenic process.


Subject(s)
Models, Animal , Nerve Regeneration/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology , Zebrafish/physiology , Animal Structures/cytology , Animal Structures/innervation , Animal Structures/physiology , Animals , Biomarkers , Cell Division/physiology
16.
Gene Expr Patterns ; 6(8): 835-42, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16545988

ABSTRACT

The secreted frizzled-related proteins (Sfrp) are a family of soluble proteins with diverse biological functions having the capacity to bind Wnt ligands, to modulate Wnt signalling, and to signal directly via the Wnt receptor, Frizzled. In an enhancer trap screen for embryonic expression in zebrafish we identified an sfrp1 gene. Previous studies suggest an important role for sfrp1 in eye development, however, no data have been reported using the zebrafish model. In this paper, we describe duplicate sfrp1 genes in zebrafish and present a detailed analysis of the expression profile of both genes. Whole mount in situ hybridisation analyses of sfrp1a during embryonic and larval development revealed a dynamic expression profile, including: the central nervous system, where sfrp1a was regionally expressed throughout the brain and developing eye; the posterior gut, from the time of endodermal cell condensation; the lateral line, where sfrp1a was expressed in the migrating primordia and interneuromast cells that give rise to the sensory organs. Other sites included the blastoderm, segmenting mesoderm, olfactory placode, developing ear, pronephros and fin-bud. We have also analysed sfrp1b expression during embryonic development. Surprisingly this gene exhibited a divergent expression profile being limited to the yolk syncytium under the elongating tail-bud, which later covered the distal yolk extension, and transiently in the tail-bud mesenchyme. Overall, our studies provide a basis for future analyses of these developmentally important factors using the zebrafish model.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Gastrula/metabolism , Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Cleavage Stage, Ovum/metabolism , Embryo, Nonmammalian , Eye/embryology , Eye/metabolism , Gene Duplication , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Mesoderm/metabolism , Molecular Sequence Data , Phylogeny , Proteins/metabolism , Sequence Homology, Amino Acid , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
17.
FEBS J ; 272(13): 3424-40, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15978047

ABSTRACT

The transforming growth factor beta (TGF-beta) superfamily includes bone morphogenetic proteins, activins and TGF-betasensu stricto (s.s). These ligands, which transduce their signal through a heteromeric complex of type I and type II receptors, have been shown to play a key role in numerous biological processes including early embryonic development in both deuterostomes and ecdyzozoans. Lophochotrozoans, the third major group of bilaterian animals, have remained in the background of the molecular survey of metazoan development. We report the cloning and functional study of the central part of the BMP pathway machinery in the bivalve mollusc Crassostrea gigas (Cg-BMPR1 type I receptor and Cg-TGFbetasfR2 type II receptor), showing an unusual functional mode of signal transduction for this superfamily. The use of the zebrafish embryo as a reporter organism revealed that Cg-BMPR1, Cg-TGFbetasfR2, Cg-ALR I, an activin Type I receptor or their dominant negative acting truncated forms, when overexpressed during gastrulation, resulted in a range of phenotypes displaying severe disturbance of anterioposterior patterning, due to strong modulations of ventrolateral mesoderm patterning. The results suggest that Cg-BMPR1, and to a certain degree Cg-TGFbetasfR2 proteins, function in C. gigas in a similar way to their zebrafish orthologues. Finally, based on phylogenetic analyses, we propose an evolutionary model within the complete TGF-beta superfamily. Thus, evidence provided by this study argues for a possible conserved endomesoderm/ectomesoderm inductive mechanism in spiralians through an ancestral BMP/activin pathway in which the singular, promiscuous and probably unique Cg-TGFbetasfR2 would be the shared type II receptor interface for both BMP and activin ligands.


Subject(s)
Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Ostreidae/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Animals , Biological Evolution , Body Patterning , Bone Morphogenetic Protein Receptors, Type I , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Dominant , Larva/cytology , Larva/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ostreidae/embryology , Ostreidae/metabolism , Phenotype , Phylogeny , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Growth Factor/genetics , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Zebrafish/embryology , Zebrafish/metabolism
18.
Development ; 131(13): 3121-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15175248

ABSTRACT

Early brain regionalisation involves the activation of genes coding for transcription factors in distinct domains of the neural plate. The limits of these domains often prefigure morphological boundaries. In the hindbrain, anteroposterior patterning depends on a segmentation process that leads to the formation of seven bulges called rhombomeres (r). The molecular cues involved in the early subdivision of the hindbrain and in rhombomere formation are not well understood. We show that iro7, a zebrafish gene coding for a transcription factor of the Iroquois family, is expressed at the end of gastrulation in the future midbrain and hindbrain territories up to the prospective r4/r5 boundary. This territory is strictly complementary to the expression domain of another homeobox gene, vhnf1, in the caudal neural plate. We demonstrate that Iro7 represses vhnf1 expression anterior to their common border and that, conversely, vHnf1 represses iro7 expression caudal to it. This suggests that the r4/r5 boundary is positioned by mutual repression between these two transcription factors. In addition, iro7 is involved in the specification of primary neurons in the rostral hindbrain. In particular, it is essential for the formation of the Mauthner neurons in r4. We propose that iro7 has a dual function in the hindbrain of the zebrafish embryo: it is required for the proper positioning of the prospective r4/r5 boundary and it promotes neurogenesis in the anterior hindbrain.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Neurons/metabolism , Rhombencephalon/embryology , Zebrafish Proteins/physiology , Animals , DNA, Complementary/metabolism , DNA-Binding Proteins/biosynthesis , Gene Library , Hepatocyte Nuclear Factor 1-beta , Immunohistochemistry , In Situ Hybridization , Protein Biosynthesis , RNA/metabolism , Signal Transduction , Time Factors , Transcription Factors/biosynthesis , Transcription, Genetic , Zebrafish
19.
Med Sci (Paris) ; 20(5): 557-61, 2004 May.
Article in French | MEDLINE | ID: mdl-15190475

ABSTRACT

Endothelial cells (EC) of the vertebrate cardiovascular system (CVS) are bona fide, yet enigmatic mechanoreceptors. When cultured in vitro and exposed to fluid forces, EC modify their physiological behaviour at the structural, metabolical and gene expression levels in response to the mechanical stimulus. However, as a direct consequence of the hypoxic bias (and often the lethality) that results from blocking blood flow in most animal systems, the in vivo role of EC mechanosensation (ECMS) remains poorly understood. The zebrafish has recently emerged as an alternative genetic model for the study of vertebrate development. Its striking ability to survive until larval stages in the absence of blood circulation circumveys the usual caveats that are inherent to CVS research, and offers the exciting opportunity to dissect the function of ECMS in vivo. Two groups have already uncovered an essential role for ECMS in zebrafish organogenesis, particularly in heart morphogenesis. In embryos in which intracardiac blood flow is genetically or physically compromised, several features of the normally developing heart, including valve formation, are specifically disrupted. In addition, impressive imaging studies of zebrafish hemodynamics demonstrate that the shear stress exerted upon the cardiac endothelium is largely in the range of the stimulus that in vitro activates cytoskelettal remodeling and gene expression changes in EC. Hence the cardiac phenotypes observed in vivo may indeed directly result from a lack of ECMS-dependent EC activity. These data shed first light on the role of ECMS in vivo. Notably, they also suggest that a number of human congenital cardiomyopathies may arise through abnormal fetal hemodynamics and/or EC sensory activity. Finally, these discoveries reinforce the too often neglected role of epigenetic factors (in this case, fluid forces) in the regulation of animal development.


Subject(s)
Hemodynamics , Mechanotransduction, Cellular/physiology , Organogenesis/physiology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Heart/embryology , Kidney/enzymology
20.
Development ; 131(3): 629-41, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14711879

ABSTRACT

Interactions between Nodal/Activin and Fibroblast growth factor (Fgf) signalling pathways have long been thought to play an important role in mesoderm formation. However, the molecular and cellular processes underlying these interactions have remained elusive. Here, we address the epistatic relationships between Nodal and Fgf pathways during early embryogenesis in zebrafish. First, we find that Fgf signalling is required downstream of Nodal signals for inducing the Nodal co-factor One-eyed-pinhead (Oep). Thus, Fgf is likely to be involved in the amplification and propagation of Nodal signalling during early embryonic stages. This could account for the previously described ability of Fgf to render cells competent to respond to Nodal/Activin signals. In addition, overexpression data shows that Fgf8 and Fgf3 can take part in this process. Second, combining zygotic mutations in ace/fgf8 and oep disrupts mesoderm formation, a phenotype that is not produced by either mutation alone and is consistent with our model of an interdependence of Fgf8 and Nodal pathways through the genetic regulation of the Nodal co-factor Oep and the cell propagation of Nodal signalling. Moreover, mesodermal cell populations are affected differentially by double loss-of-function of Zoep;ace. Most of the dorsal mesoderm undergoes massive cell death by the end of gastrulation, in contrast to either single-mutant phenotype. However, some mesoderm cells are still able to undergo myogenic differentiation in the anterior trunk of Zoep;ace embryos, revealing a morphological transition at the level of somites 6-8. Further decreasing Oep levels by removing maternal oep products aggravates the mesodermal defects in double mutants by disrupting the fate of the entire mesoderm. Together, these results demonstrate synergy between oep and fgf8 that operates with regional differences and is involved in the induction, maintenance, movement and survival of mesodermal cell populations.


Subject(s)
Embryo, Nonmammalian/metabolism , Fibroblast Growth Factors/metabolism , Mesoderm/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation/physiology , Zebrafish Proteins , Animals , Apoptosis/physiology , Body Patterning/physiology , Fibroblast Growth Factor 3 , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/genetics , Gastrula/physiology , Nodal Protein , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Zebrafish/embryology , Zebrafish/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...