Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Opt Express ; 29(6): 9098-9122, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33820345

ABSTRACT

We analytically and numerically investigate surface second-harmonic generation (SHG) from a stack of dielectric layers. We develop a theoretical formalism based on the transfer matrix method for the calculation of the surface-driven second-harmonic radiation from multilayer structures and elaborate it for the case of ultrathin dielectric layers using a power series expansion to derive the effective surface nonlinear tensor for the whole stack. We show that for deeply subwavelength thicknesses of the layers the surface responses from all interfaces can efficiently sum up, leading to largely enhanced efficiency of SHG. As a result, such surface-driven nonlinearity can become comparable to the bulk nonlinearity in noncentrosymmetric semiconductors and can yield high performance for nonlinear nanophotonic applications.

2.
Sci Rep ; 10(1): 10545, 2020 Jun 29.
Article in English | MEDLINE | ID: mdl-32601374

ABSTRACT

We analyse possibilities to quantitatively evaluate the surface second-order optical nonlinearity in noncentrosymmetric materials based on polarization-resolved analysis of far-field radiation patterns of second-harmonic generation. We analytically demonstrate that for plane-wave illumination the contribution to the second-harmonic signal from the surface of a nonlinear medium exhibits different polarization properties and angular dependencies compared to the contribution from the bulk. In view of this, we optimize the illumination geometry in order to enable the most efficient separation and comparison of both nonlinearities. Furthermore, we consider the illumination of an AlGaAs slab by a tightly-focused linearly-polarized Gaussian beam as an alternative measurement geometry. It is found that the reliable separation of the surface nonlinearity contribution as well as a wide range of detectable values can be achieved with this geometry as well.

3.
Sci Rep ; 4: 5886, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25073935

ABSTRACT

Crystalline silicon photonic crystal slabs are widely used in various photonics applications. So far, the commercial success of such structures is still limited owing to the lack of cost-effective fabrication processes enabling large nanopatterned areas (≫ 1 cm(2)). We present a simple method for producing crystalline silicon nanohole arrays of up to 5 × 5 cm(2) size with lattice pitches between 600 and 1000 nm on glass and flexible plastic substrates. Exclusively up-scalable, fast fabrication processes are applied such as nanoimprint-lithography and silicon evaporation. The broadband light trapping efficiency of the arrays is among the best values reported for large-area experimental crystalline silicon nanostructures. Further, measured photonic crystal resonance modes are in good accordance with light scattering simulations predicting strong near-field intensity enhancements greater than 500. Hence, the large-area silicon nanohole arrays might become a promising platform for ultrathin solar cells on lightweight substrates, high-sensitive optical biosensors, and nonlinear optics.

4.
Cell Death Differ ; 13(2): 223-35, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16096653

ABSTRACT

Cell culture work has identified the tumor suppressor p53 as a component of the S-phase checkpoint control system, while in vivo studies of this role of p53 in whole-vertebrate systems were limited. Here, we describe zebrafish mutants in the DNA polymerase delta catalytic subunit 1, based on the positional cloning of the flathead (fla) gene. fla mutants display specific defects in late proliferative zones, such as eyes, brain and cartilaginous elements of the visceral head skeleton, where cells display compromised DNA replication, followed by apoptosis, and partial or complete loss of affected tissues. Antisense-mediated knockdown of p53 in fla mutants leads to a striking rescue of all phenotypic traits, including completion of replication, survival of cells, and normal differentiation and tissue formation. This indicates that under replication-compromised conditions, the p53 branch of the S-phase checkpoint is responsible for eliminating stalled cells that, given more time, would have otherwise finished their normal developmental program.


Subject(s)
Apoptosis , Cell Differentiation , DNA Polymerase III/metabolism , Mutation , Tumor Suppressor Protein p53/physiology , Zebrafish/genetics , Amino Acid Sequence , Animals , Brain/cytology , Brain/embryology , Cell Proliferation , DNA Polymerase III/deficiency , DNA Polymerase III/genetics , DNA Replication , Eye/cytology , Eye/embryology , Female , Gene Expression Regulation, Developmental , Male , Molecular Sequence Data , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Skull/cytology , Skull/embryology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics , Up-Regulation/physiology , Zebrafish/embryology , Zebrafish/physiology , Zygote/cytology , Zygote/enzymology , Zygote/physiology
5.
Cell Death Differ ; 12(1): 52-64, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15529176

ABSTRACT

The tumor suppressor p53 has two alternative effects, causing either cell cycle arrest or apoptosis. These different effects are supposed to be mediated by the transcriptional activation of different target genes. perp, encoding a transmembrane protein of the Pmp22 family, is a transcriptional p53 target exclusively upregulated in apoptotic cells. However, its role during normal development had remained largely unclear. Here, we report the isolation and characterization of a zebrafish perp homolog. Upon overexpression in early zebrafish embryos, perp induces apoptosis. In addition, it contributes to p53-dependent and UV-induced cell death. However, during normal zebrafish development, perp displays a p53-independent and spatially restricted expression in specific cell types and tissues. Antisense-mediated loss of Perp function leads to increased apoptosis in perp-expressing cells of the developing skin and notochord. We conclude that, in contrast to its proapoptotic function in stressed cells, Perp plays an antiapoptotic role during normal zebrafish development to regulate tissue-specific cell survival.


Subject(s)
Membrane Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Caspase Inhibitors , Caspases/metabolism , Cell Survival/radiation effects , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/radiation effects , Gene Expression Regulation, Developmental/radiation effects , Membrane Proteins/physiology , Notochord/embryology , Notochord/metabolism , Phosphoproteins/genetics , RNA, Messenger/administration & dosage , Skin/embryology , Skin/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays , Zebrafish/genetics , Zebrafish Proteins/physiology
6.
Dev Dyn ; 222(4): 681-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11748836

ABSTRACT

Bone morphogenetic proteins (Bmps) and their roles during early dorsoventral patterning of the vertebrate embryo are well understood. The role and regulation of a more distant member of this family, the anti-dorsalizing morphogenetic protein (Admp), however, are less clear. Here, we report the isolation and characterization of zebrafish admp. Unlike other bmps, admp is exclusively expressed on the dorsal side. Expression starts at blastula stages in the region of the organizer, giving rise to anterior neuroectoderm and axial mesoderm. During the course of gastrulation, both the neuroectodermal and the mesodermal admp transcripts vanish in an anterior-posterior wave. The maintenance of admp expression is positively influenced by Nodal signaling and by Bozozok (Boz), an organizer-promoting homeodomain protein acting as a repressor of early bmp2b expression. Despite the positive effect of boz on admp expression, Boz and Admp have rather opposite effects on zebrafish patterning, as revealed in gain- and loss-of-function experiments. Upon overexpression, admp has Bmp-like activities causing a smaller organizer and enhanced ventral specification, very similar to the phenotype caused by the loss of boz function in mutant embryos. Antisense-based admp knockdown, on the other side, leads to an enlarged organizer and impaired ventral and posterior development, as observed in embryos after boz overexpression. This finding indicates that admp is required for the development of embryonic structures normally suppressed by organizer activities. The seeming discrepancy between the regulative and functional relationship of boz and admp is discussed, and models are proposed according to which Admp might be part of a negative feedback loop to pattern and confine the organizer region.


Subject(s)
Bone Morphogenetic Proteins/physiology , Organizers, Embryonic/physiology , Zebrafish Proteins , Zebrafish/embryology , Animals , Blastocyst/physiology , Body Patterning/physiology , Bone Morphogenetic Proteins/isolation & purification , Bone Morphogenetic Proteins/metabolism , Embryo, Nonmammalian/physiology , Gastrula/physiology , Homeodomain Proteins/physiology , Mesoderm/physiology , Nervous System/embryology , Nodal Protein , Signal Transduction/physiology , Tissue Distribution , Transforming Growth Factor beta/physiology
7.
Dev Cell ; 1(2): 251-64, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11702784

ABSTRACT

Mutations in the zebrafish knypek locus impair gastrulation movements of convergent extension that narrow embryonic body and elongate it from head to tail. We demonstrate that knypek regulates cellular movements but not cell fate specification. Convergent extension movement defects in knypek are associated with abnormal cell polarity, as mutant cells fail to elongate and align medio-laterally. Positional cloning reveals that knypek encodes a member of the glypican family of heparan sulfate proteoglycans. Double mutant and overexpression analyses show that Knypek potentiates Wnt11 signaling, mediating convergent extension. These studies provide experimental and genetic evidence that glypican Knypek acts during vertebrate gastrulation as a positive modulator of noncanonical Wnt signaling to establish polarized cell behaviors underlying convergent extension movements.


Subject(s)
Gastrula/physiology , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/physiology , Zebrafish Proteins , Amino Acid Sequence , Animals , Body Patterning , Cell Division , Cloning, Molecular , Cysteine/chemistry , Dose-Response Relationship, Drug , Glycoproteins/metabolism , In Situ Hybridization , Models, Genetic , Molecular Sequence Data , Mutation , Phenotype , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Wnt Proteins , Zebrafish
8.
Mech Dev ; 108(1-2): 179-84, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11578872

ABSTRACT

In this paper we describe the mRNA expression patterns of members of the bone morphogenetic protein (BMP) signalling pathway in the developing zebrafish ear. bmp2b, 4, and 7 are expressed in discrete areas of otic epithelium, some of which correspond to sensory patches. bmp2b and 4 mark the developing cristae before and during the appearance of differentiated hair cells. bmp4 is also expressed in a dorsal, non-sensory region of the ear. Expression of bmps in cristae is conserved between zebrafish, chick, and mouse, but there are also notable differences in ear expression patterns between these species. Of five zebrafish BMP antagonists, only one (follistatin) shows significant expression in the otic epithelium. The type I receptor bmpr-IB shows localised expression in the ear epithelium. Mediators of BMP signalling, smad1 and smad5, are expressed in statoacoustic and lateral line ganglia; smad5 is also expressed at low levels throughout the ear epithelium. An inhibitory smad, smad6, is expressed laterally in the ear epithelium. Lateral line primordia and neuromasts also express bmp2b, 4, follistatin, smad1, and smad5. The conservation of bmp expression in cristae among different species adds weight to the growing evidence that BMPs are required for the development of the vertebrate ear.


Subject(s)
Bone Morphogenetic Proteins/genetics , Ear, Inner/embryology , Zebrafish/embryology , Zebrafish/genetics , Animals , Body Patterning/genetics , Chick Embryo , Gene Expression Regulation, Developmental , In Situ Hybridization , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
10.
Development ; 128(6): 849-58, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222140

ABSTRACT

Ventral specification of mesoderm and ectoderm depends on signaling by members of the bone morphogenetic protein (Bmp) family. Bmp signals are transmitted by a complex of type I and type II serine/threonine kinase transmembrane receptors. Here, we show that Alk8, a novel member of the Alk1 subgroup of type I receptors, is disrupted in zebrafish lost-a-fin (laf) mutants. Two alk8/laf null alleles are described. In laf(tm110), a conserved extracellular cysteine residue is replaced by an arginine, while in laf(m100), Alk8 is prematurely terminated directly after the transmembrane domain. The zygotic effect of both mutations leads to dorsalization of intermediate strength. A much stronger dorsalization, similar to that of bmp2b/swirl and bmp7/snailhouse mutants, however, is obtained by inhibiting both maternally and zygotically supplied alk8 gene products with morpholino antisense oligonucleotides. The phenotype of laf mutants and alk8 morphants can be rescued by injected mRNA encoding Alk8 or the Bmp-regulated transcription factor Smad5, but not by mRNA encoding Bmp2b or Bmp7. Conversely, injected mRNA encoding a constitutively active version of Alk8 can rescue the strong dorsalization of bmp2b/swirl and bmp7/snailhouse mutants, whereas smad5/somitabun mutant embryos do not respond. Altogether, the data suggest that Alk8 acts as a Bmp2b/7 receptor upstream of Smad5.


Subject(s)
Body Patterning/physiology , Bone Morphogenetic Proteins/physiology , Embryo, Nonmammalian/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Transforming Growth Factor beta , Zebrafish Proteins , Zebrafish/embryology , Activin Receptors , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 7 , Cloning, Molecular , Crosses, Genetic , Genetic Linkage , Genotype , Mutagenesis , Phenotype , Phylogeny , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Zebrafish/genetics
13.
Gene ; 246(1-2): 69-80, 2000 Apr 04.
Article in English | MEDLINE | ID: mdl-10767528

ABSTRACT

smad genes encode transcription factors involved in the signal transduction of members of the TGFbeta superfamily. We report here the cloning, characterization and genomic mapping of smad2, smad3 and smad4 from the zebrafish, Danio rerio. In Xenopus, smad2 overexpression has been shown to interfere with gastrulation and dorsal cell fate specification. However, full-length zebrafish smad2, although functionally active in Xenopus explants, has no effect when overexpressed in zebrafish embryos. In contrast, an N-terminally truncated, constitutively active version of Smad2 protein causes severe dorsalization or partial secondary axis formation, pointing to a role of Smad2 during mesoderm and axis formation. The temporal and spatial expression patterns of zebrafish smad2, 3 and 4 were investigated by developmental RT-PCR and whole mount in-situ hybridization. All three genes show strong and ubiquitous maternal expression. Zygotic expression is weak and ubiquitous in the case of smad2, and strong and ubiquitious in the case of smad4, while smad3 shows a spatially restricted zygotic expression pattern. It is expressed in migrating neural crest cells of the trunk and a subset of cells in the diencephalon in close proximity to the expression domain of the Nodal-related protein Cyclops/Ndr2/Znr1, a potential signal upstream of Smad2/3 required for eye-field separation and floor plate specification. Overexpression of truncated smad2 in cyclops mutant embryos leads to a rescue of the eye and floorplate defects. These data suggest that Smad2 acts as a mediator of Nodal signals during zebrafish midline signaling, while Smad3 might be involved in later steps of eye field separation.


Subject(s)
DNA-Binding Proteins/genetics , Trans-Activators/genetics , Xenopus Proteins , Zebrafish Proteins , Zebrafish/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Embryonic Development , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutation , Nerve Growth Factors , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Smad Proteins , Smad2 Protein , Smad3 Protein , Smad4 Protein , Transforming Growth Factor beta/genetics , Xenopus/genetics , Zebrafish/embryology
14.
Development ; 127(2): 343-54, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10603351

ABSTRACT

Bone morphogenetic proteins (Bmps) are signaling molecules that have been implicated in a variety of inductive processes. We report here that zebrafish Bmp7 is disrupted in snailhouse (snh) mutants. The allele snh(st1) is a translocation deleting the bmp7 gene, while snh(ty68) displays a Val->Gly exhange in a conserved motif of the Bmp7 prodomain. The snh(ty68) mutation is temperature-sensitive, leading to severalfold reduced activity of mutant Bmp7 at 28 degrees C and non-detectable activity at 33 degrees C. This prodomain lesion affects secretion and/or stability of secreted mature Bmp7 after processing has occurred. Both snh(st1) and snh(ty68) mutant zebrafish embryos are strongly dorsalized, indicating that bmp7 is required for the specification of ventral cell fates during early dorsoventral patterning. At higher temperature, the phenotype of snh(ty68) mutant embryos is identical to that caused by the amorphic bmp2b mutation swirl swr(ta72) and similar to that caused by the smad5 mutation somitabun sbn(dtc24). mRNA injection studies and double mutant analyses indicate that Bmp2b and Bmp7 closely cooperate and that Bmp2b/Bmp7 signaling is transduced by Smad5 and antagonized by Chordino.


Subject(s)
Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Transforming Growth Factor beta , Zebrafish/embryology , Amino Acid Sequence , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Cell Transplantation , Cloning, Molecular , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Glycoproteins/genetics , In Situ Hybridization , Molecular Sequence Data , Mutation , Phosphoproteins/genetics , RNA, Messenger/metabolism , Sequence Alignment , Signal Transduction , Smad5 Protein , Trans-Activators/genetics , Zebrafish/genetics , Zebrafish Proteins
15.
Dev Dyn ; 216(3): 285-98, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10590480

ABSTRACT

Smad1 and smad5 encode transcription factors that have been implicated in the transduction of signaling by the bone morphogenetic proteins Bmp2 and/or Bmp4. Here we report the characterization of Smad1 and Smad5 from the zebrafish, Danio rerio. Although smad1, smad5, bmp2b, and bmp4 are all expressed during gastrulation and although all four proteins have ventralizing activities, they appear to play distinct roles during dorsoventral pattern formation. smad1 expression starts shortly before the onset of gastrulation. It is expressed on the ventral side of the embryo, whereas smad5 transcripts are both maternally and zygotically provided and ubiquitously distributed. Injection studies and mutant analyses suggest that the ventral smad1 expression is positively regulated by Bmp2b, but not by Bmp4 signaling, whereas smad5 expression is independent of Bmp2b. Also, the dorsalized phenotype of bmp2b-mutant embryos can be rescued by exogenous Smad1, but not by Smad5. Together, these data suggest that smad1 acts later than smad5 and is itself a transcriptional target of Smad5-mediated Bmp2b signaling. During later stages of development, smad1 is expressed in eyes, dorsal cells of rhombomeres 1, 3, and 5, and somites, with highest mRNA levels in the presumptive sclerotome and adaxial regions near the notochord. Injection experiments indicate that this somitic smad1 expression is positively regulated by hedgehog signaling from the dorsal midline, thus perhaps accounting for the recently reported sonic hedgehog-induced competence of sclerotomal cells to Bmp2/4 signals.


Subject(s)
Body Patterning/physiology , DNA-Binding Proteins/metabolism , Drosophila Proteins , Phosphoproteins/metabolism , Trans-Activators/metabolism , Zebrafish/embryology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins , In Situ Hybridization , Insect Proteins/metabolism , Mutation , Phenotype , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Smad Proteins , Smad5 Protein , Time Factors , Tissue Distribution , Transforming Growth Factor beta/metabolism , Zebrafish Proteins
16.
Genes Dev ; 13(16): 2072-86, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10465785

ABSTRACT

The mechanisms that control cell proliferation and cell differentiation during morphogenesis of the endochondral skeleton of vertebrates are poorly understood. Indian hedgehog (Ihh) signaling from prehypertrophic chondrocytes has been implicated in the control of chondrocyte maturation by way of feedback control of a second secreted factor parathyroid hormone-related peptide (PTHrP) at the articular surfaces. Analysis of an Ihh null mutant suggests a more extensive role for Ihh in skeletal development. Mutants display markedly reduced chondrocyte proliferation, maturation of chondrocytes at inappropriate position, and a failure of osteoblast development in endochondral bones. Together, the results suggest a model in which Ihh coordinates diverse aspects of skeletal morphogenesis through PTHrP-dependent and independent processes.


Subject(s)
Chondrocytes/cytology , Osteogenesis/physiology , Proteins/metabolism , Signal Transduction , Trans-Activators , Alleles , Animals , Cartilage/cytology , Cell Differentiation , Cell Division , Gene Deletion , Hedgehog Proteins , Mice , Mice, Inbred C57BL , Osteoblasts/physiology , Proteins/genetics
17.
Development ; 126(10): 2149-59, 1999 May.
Article in English | MEDLINE | ID: mdl-10207140

ABSTRACT

Signaling by members of the TGFbeta superfamily is thought to be transduced by Smad proteins. Here, we describe a zebrafish mutant in smad5, designated somitabun (sbn). The dominant maternal and zygotic effect of the sbntc24 mutation is caused by a change in a single amino acid in the L3 loop of Smad5 protein which transforms Smad5 into an antimorphic version, inhibiting wild-type Smad5 and related Smad proteins. sbn mutant embryos are strongly dorsalized, similarly to mutants in Bmp2b, its putative upstream signal. Double mutant analyses and RNA injection experiments show that sbn and bmp2b interact and that sbn acts downstream of Bmp2b signaling to mediate Bmp2b autoregulation during early dorsoventral (D-V) pattern formation. Comparison of early marker gene expression patterns, chimera analyses and rescue experiments involving temporally controlled misexpression of bmp or smad in mutant embryos reveal three phases of D-V patterning: an early sbn- and bmp2b-independent phase when a coarse initial D-V pattern is set up, an intermediate sbn- and bmp2b-dependent phase during which the putative morphogenetic Bmp2/4 gradient is established, and a later sbn-independent phase during gastrulation when the Bmp2/4 gradient is interpreted and cell fates are specified.


Subject(s)
Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transforming Growth Factor beta , Amino Acid Sequence , Animals , Base Sequence , Body Patterning , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , DNA, Complementary , DNA-Binding Proteins/genetics , Genetic Linkage , Humans , Molecular Sequence Data , Phenotype , Phosphoproteins/genetics , Smad Proteins , Smad5 Protein , Trans-Activators/genetics , Zebrafish , Zebrafish Proteins
19.
Dev Genet ; 23(2): 97-103, 1998.
Article in English | MEDLINE | ID: mdl-9770266

ABSTRACT

The BMP family of polypeptide growth factors has been shown to play diverse roles in establishing embryonic patterning and tissue fates. We report the cloning of the zebrafish homologue of BMP-2, examine its expression during embryogenesis, and find that it is localized to the distal end of the long arm of zebrafish chromosome 20. A missense mutation of the bmp2 gene has recently been shown to be responsible for the early dorsalized phenotype of the zebrafish swirl mutant [Kishimoto et al., 1997]. Given the dynamic expression of bmp2 in the developing embryo and the complex interactions of BMP signaling response in vertebrates, it is possible that other mutant phenotypes, due to altered bmp2 gene expression, will eventually map to or interact with this genetic locus.


Subject(s)
Bone Morphogenetic Proteins/genetics , Genes , Transforming Growth Factor beta , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/biosynthesis , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Gene Expression Regulation, Developmental , Molecular Sequence Data , Mutation, Missense , Sequence Alignment , Sequence Homology, Amino Acid , Zebrafish/embryology
20.
Dev Biol ; 194(2): 166-71, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9501021

ABSTRACT

Recent results have indicated that cAMP-dependent protein kinase (PKA) acts as a negative regulator of Hedgehog signaling in target cells of the vertebrate embryo. Consequently, suppression of PKA activity is sufficient to mimic the effect of receiving a Hedgehog signal. We have explored whether PKA-inhibiting Gi-proteins (GiPs) may also be involved in the regulation of Hedgehog signaling. Zebrafish embryos were injected with RNA encoding pertussis toxin (Ptx), a specific inhibitor of GiPs. These embryos developed phenotypic traits opposite to embryos expressing a dominant negative form of the PKA regulatory subunit (dnPKA), including a fusion of the eyes, a lack of ventral specification in the forebrain, and an expansion of the sclerotome at the expense of adaxial fates in the posterior somites. These effects can be partially rescued by coexpression of dnPKA, but not by coexpression of Indian Hedgehog, suggesting that GiPs act upstream of PKA and downstream of Hedgehogs. Other Hedgehog- and PKA-dependent processes, sclerotomal specification and adaxial specification in the first five somites, are not negatively affected by Ptx. Thus, GiPs may be involved in Hedgehog signaling in some, but not all target cells.


Subject(s)
Embryonic Induction , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Pertussis Toxin , Proteins/physiology , Signal Transduction , Trans-Activators , Virulence Factors, Bordetella/metabolism , Zebrafish/embryology , Animals , Body Patterning , Brain/embryology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dimerization , Eye/embryology , Gene Transfer Techniques , Hedgehog Proteins , Ligands , RNA/metabolism , Receptors, Cell Surface/metabolism , Virulence Factors, Bordetella/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...