Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Nature ; 613(7942): 153-159, 2023 01.
Article in English | MEDLINE | ID: mdl-36517597

ABSTRACT

Sequential segmentation creates modular body plans of diverse metazoan embryos1-4. Somitogenesis establishes the segmental pattern of the vertebrate body axis. A molecular segmentation clock in the presomitic mesoderm sets the pace of somite formation4. However, how cells are primed to form a segment boundary at a specific location remains unclear. Here we developed precise reporters for the clock and double-phosphorylated Erk (ppErk) gradient in zebrafish. We show that the Her1-Her7 oscillator drives segmental commitment by periodically lowering ppErk, therefore projecting its oscillation onto the ppErk gradient. Pulsatile inhibition of the ppErk gradient can fully substitute for the role of the clock, and kinematic clock waves are dispensable for sequential segmentation. The clock functions upstream of ppErk, which in turn enables neighbouring cells to discretely establish somite boundaries in zebrafish5. Molecularly divergent clocks and morphogen gradients were identified in sequentially segmenting species3,4,6-8. Our findings imply that versatile clocks may establish sequential segmentation in diverse species provided that they inhibit gradients.


Subject(s)
Body Patterning , Extracellular Signal-Regulated MAP Kinases , Periodicity , Somites , Zebrafish Proteins , Zebrafish , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Somites/drug effects , Somites/embryology , Somites/enzymology , Somites/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism , Biological Clocks , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism
2.
J Anat ; 236(4): 622-629, 2020 04.
Article in English | MEDLINE | ID: mdl-31840255

ABSTRACT

In the vertebrate body, a metameric structure is present along the anterior-posterior axis. Zebrafish tbx6-/- larvae, in which somite boundaries do not form during embryogenesis, were shown to exhibit abnormal skeletal morphology such as rib, neural arch and hemal arch. In this study, we investigated the role of somite patterning in the formation of anterior vertebrae and ribs in more detail. Using three-dimensional computed tomography scans, we found that anterior vertebrae including the Weberian apparatus were severely affected in tbx6-/- larvae. In addition, pleural ribs of tbx6 mutants exhibited severe defects in the initial ossification, extension of ossification, and formation of parapophyses. Two-colour staining revealed that bifurcation of ribs was caused by fusion or branching of ribs in tbx6-/- . The parapophyses in tbx6-/- juvenile fish showed irregular positioning to centra and abnormal attachment to ribs. Furthermore, we found that the ossification of the distal portion of ribs proceeded along myotome boundaries even in irregularly positioned myotome boundaries. These results provide evidence of the contribution of somite patterning to the formation of the Weberian apparatus and rib in zebrafish.


Subject(s)
Body Patterning/genetics , Ribs/embryology , Somites/enzymology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Gene Expression Regulation, Developmental , Ribs/diagnostic imaging , Somites/diagnostic imaging , T-Box Domain Proteins/genetics , Tomography, X-Ray Computed , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Sci Rep ; 8(1): 4335, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29531317

ABSTRACT

During somite segmentation, clock genes oscillate within the posterior presomitic mesoderm (PSM). The temporal information ties up with the posteriorly moving FGF gradient, leading to the formation of a presumptive somite within the PSM. We previously investigated Erk activity downstream of FGF signaling by collecting stained zebrafish embryos, and discovered that the steep gradient of Erk activity was generated in the PSM, and the Erk activity border regularly shifted in a stepwise manner. However, since these interpretations come from static analyses, we needed to firmly confirm them by applying an analysis that has higher spatiotemporal resolutions. Here we developed a live imaging system for Erk activity in zebrafish embryos, using a Förster resonance energy transfer (FRET)-based Erk biosensor. With this system, we firmly showed that Erk activity exhibits stepwise regression within the PSM. Although our static analyses could not detect the stepwise pattern of Erk activity in clock-deficient embryos, our system revealed that, in clock-deficient embryos, the stepwise regression of Erk activity occurs at an irregular timing, eventually leading to formation of irregularly-sized somites. Therefore, our system overcame the limitation of static analyses and revealed that clock-dependent spatiotemporal regulation of Erk is required for proper somitogenesis in zebrafish.


Subject(s)
MAP Kinase Signaling System , Somites/enzymology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Biosensing Techniques/methods , Body Patterning , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/ultrastructure , Embryonic Development , Fluorescence Resonance Energy Transfer/methods , Gene Expression Regulation, Developmental , Zebrafish Proteins/genetics
4.
Int J Dev Biol ; 61(6-7): 459-463, 2017.
Article in English | MEDLINE | ID: mdl-28695966

ABSTRACT

Dual specificity tyrosine-phosphorylation regulated kinase 2 (DYRK2) is a serine/threonine kinase. In zebrafish, DYRK2 is expressed in the lateral somites and adaxial cells at the early stage of embryo development. However, its role in early myogenesis had not been elucidated yet. Here, we report that DYRK2 mRNA and MyoD mRNA were colocalized in the muscle progenitor cells in somites, including both the posterior compartment of the lateral somites and adaxial cells. Knockdown of DYRK2 reduced the levels of MyoD transcripts in the muscle progenitor cells in somites. In contrast, overexpression of DYRK2 increased the levels of MyoD transcripts in the muscle progenitor cells in somites. The effects of knockdown and overexpression of DYRK2 on the expression of MyoD in the posterior compartment of the lateral somites were much greater than in the adaxial cells. Further studies indicated that forced expression of DYRK2 increased the levels of fast-twitch skeletal myosin RNA. Moreover, knockdown or forced expression of DYRK2 affected the levels of fast-twitch skeletal myosin protein. Together, these data indicate that DYRK2 is expressed in the developing muscle progenitor cells in somites and that it positively regulates fast-twitch muscle differentiation, at least at the early stages.


Subject(s)
Embryo, Nonmammalian/cytology , Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/cytology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Somites/growth & development , Zebrafish/growth & development , Animals , Cell Differentiation , Embryo, Nonmammalian/metabolism , Muscle Development/physiology , Muscle Fibers, Skeletal/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Somites/enzymology , Zebrafish/genetics , Zebrafish/metabolism , Dyrk Kinases
5.
FEBS J ; 281(19): 4367-83, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25056693

ABSTRACT

Glycogen synthase kinase 3 beta (Gsk3b) acts as a negative modulator in endothelial cells through the Wnt/ß-catenin/PI3K/AKT/Gsk3b axis in cancer-induced angiogenesis. However, the function of Gsk3b during embryonic angiogenesis remains unclear. Here, either gsk3b knockdown by morpholino or Gsk3b loss of activity by LiCl treatment had serious phenotypic consequences, such as defects in the positioning and patterning of intersegmental blood vessels and reduction of vegfaa121 and vegfaa165 transcripts. In embryos treated with the phosphatidylinositol 3-kinase inhibitor, angiogenesis was severely inhibited, along with reduced Wnt, phosphorylated AKT and phosphorylated Gsk3b, suggesting that the remaining Gsk3b in somites could still degrade ß-catenin, resulting in decreased vascular endothelial growth factor Aa(VegfAa) expression. However, in gsk3b-mRNA-overexpressed embryos, intersegmental vessels ectopically sprouted by the increase in phosphorylated-Gsk3b which prevented the degradation of ß-catenin and promoted the increase in phosphorylated AKT activity, thus increasing VegfAa expression in somites. Interestingly, the Gsk3b-dependent cross-talk between PI3K/AKT and Wnt/ß-catenin suggests that Wnt/ß-catenin and PI3K/AKT interaction controls embryonic angiogenesis by a positive feedback loop rather than a hierarchical framework such as that found in cancer-induced angiogenesis. Thus, both active and inactive forms of Gsk3b mediate the cooperative signaling between Wnt/ß-catenin and PI3K/AKT to control VegfAa expression in somites during angiogenesis in zebrafish embryos.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Neovascularization, Physiologic , Somites/enzymology , Zebrafish Proteins/physiology , Animals , Cleavage Stage, Ovum/enzymology , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/enzymology , Endothelium, Vascular/enzymology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3 beta , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/metabolism , Zebrafish
6.
Mol Cell Biochem ; 356(1-2): 209-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21761203

ABSTRACT

CK2 is a highly conserved serine-threonine kinase involved in biological processes such as embryonic development, circadian rhythms, inflammation, and cancer. Biochemical experiments have implicated CK2 in the control of several cellular processes and in the regulation of signal transduction pathways. Our laboratory is interested in characterizing the cellular, signaling, and molecular mechanisms regulated by CK2 during early embryonic development. For this purpose, animal models, including mice deficient in CK2 genes, are indispensable tools. Using CK2α gene-deficient mice, we have recently shown that CK2α is a critical regulator of mid-gestational morphogenetic processes, as CK2α deficiency results in defects in heart, brain, pharyngeal arch, tail bud, limb bud, and somite formation. Morphogenetic processes depend upon the precise coordination of essential cellular processes in which CK2 has been implicated, such as proliferation and survival. Here, we summarize the overall phenotype found in CK2α (-/- ) mice and describe our initial analysis aimed to identify the cellular processes affected in CK2α mutants.


Subject(s)
Casein Kinase II/metabolism , Embryonic Development , Morphogenesis , Animals , Apoptosis , Casein Kinase II/deficiency , Cell Proliferation , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/cytology , Embryo, Mammalian/enzymology , Head/embryology , Limb Buds/embryology , Mice , Phenotype , Somites/cytology , Somites/enzymology , Tail/embryology
7.
Proc Natl Acad Sci U S A ; 107(46): 19921-6, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-20980661

ABSTRACT

Inositol phosphate (IP) kinases constitute an emerging class of cellular kinases linked to multiple cellular activities. Here, we report a previously uncharacterized cellular function in Hedgehog (Hh) signaling for the IP kinase designated inositol hexakisphosphate kinase-2 (IP6K2) that produces diphosphoryl inositol phosphates (PP-IPs). In zebrafish embryos, IP6K2 activity was required for normal development of craniofacial structures, somites, and neural crest cells. ip6k2 depletion in both zebrafish and mammalian cells also inhibited Hh target gene expression. Inhibiting IP(6) kinase activity using N(2)-(m-(trifluoromethy)lbenzyl) N(6)-(p-nitrobenzyl)purine (TNP) resulted in altered Hh signal transduction. In zebrafish, restoring IP6K2 levels with exogenous ip6k2 mRNA reversed the effects of IP6K2 depletion. Furthermore, overexpression of ip6k2 in mammalian cells enhanced the Hh pathway response, suggesting IP6K2 is a positive regulator of Hh signaling. Perturbations from IP6K2 depletion or TNP were reversed by overexpressing smoM2, gli1, or ip6k2. Moreover, the inhibitory effect of cyclopamine was reversed by overexpressing ip6k2. This identified roles for the inositol kinase pathway in early vertebrate development and tissue morphogenesis, and in Hh signaling. We propose that IP6K2 activity is required at the level or downstream of Smoothened but upstream of the transcription activator Gli1.


Subject(s)
Hedgehog Proteins/metabolism , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Movement , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/enzymology , Craniofacial Abnormalities/pathology , Embryo, Mammalian/enzymology , Embryo, Mammalian/pathology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Inositol Phosphates/metabolism , Mice , NIH 3T3 Cells , Neural Crest/enzymology , Neural Crest/pathology , Somites/abnormalities , Somites/enzymology , Somites/pathology , Zebrafish/embryology , Zebrafish/genetics
8.
Dev Biol ; 347(1): 195-203, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20801112

ABSTRACT

Mice deficient in growth differentiation factor 11 (GDF11) signaling display anterior transformation of axial vertebrae and truncation of caudal vertebrae. However, the in vivo molecular mechanisms by which GDF11 signaling regulates the development of the vertebral column have yet to be determined. We found that Gdf11 and Acvr2b mutants are sensitive to exogenous RA treatment on vertebral specification and caudal vertebral development. We show that diminished expression of Cyp26a1, a retinoic acid inactivating enzyme, and concomitant elevation of retinoic acid activity in the caudal region of Gdf11(-/-) embryos may account for this phenomenon. Reduced expression or function of Cyp26a1 enhanced anterior transformation of axial vertebrae in wild-type and Acvr2b mutants. Furthermore, a pan retinoic acid receptor antagonist (AGN193109) could lessen the anterior transformation phenotype and rescue the tail truncation phenotype of Gdf11(-/-) mice. Taken together, these results suggest that GDF11 signaling regulates development of caudal vertebrae and is involved in specification of axial vertebrae in part by maintaining Cyp26a1 expression, which represses retinoic acid activity in the caudal region of embryos during the somitogenesis stage.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Signal Transduction , Spine/embryology , Spine/metabolism , Tretinoin/metabolism , Activin Receptors, Type II/metabolism , Animals , Body Patterning/drug effects , Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Embryo, Mammalian/drug effects , Embryo, Mammalian/enzymology , Gene Expression Regulation, Developmental/drug effects , Growth Differentiation Factors/genetics , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/enzymology , Mice , Mutation/genetics , Retinoic Acid 4-Hydroxylase , Signal Transduction/drug effects , Somites/drug effects , Somites/embryology , Somites/enzymology , Spine/drug effects , Tail/abnormalities , Tail/drug effects , Tretinoin/pharmacology , Wnt Proteins/metabolism , Wnt3 Protein
9.
Development ; 137(16): 2753-62, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20630945

ABSTRACT

Intracellular calcium ion (Ca(2+)) elevation on the left side of the mouse embryonic node or zebrafish Kupffer's vesicle (KV) is the earliest asymmetric molecular event that is functionally linked to lateral organ placement in these species. In this study, Ca(2+)/CaM-dependent protein kinase (CaMK-II) is identified as a necessary target of this Ca(2+) elevation in zebrafish embryos. CaMK-II is transiently activated in approximately four interconnected cells along the anterior left wall of the KV between the six- and 12-somite stages, which is coincident with known left-sided Ca(2+) elevations. Within these cells, activated CaMK-II is observed at the surface and in clusters, which appear at the base of some KV cilia. Although seven genes encode catalytically active CaMK-II in early zebrafish embryos, one of these genes also encodes a truncated inactive variant (alphaKAP) that can hetero-oligomerize with and target active enzyme to membranes. alphaKAP, beta2 CaMK-II and gamma1 CaMK-II antisense morpholino oligonucleotides, as well as KV-targeted dominant negative CaMK-II, randomize organ laterality and southpaw (spaw) expression in lateral plate mesoderm (LPM). Left-sided CaMK-II activation was most dependent on an intact KV, the PKD2 Ca(2+) channel and gamma1 CaMK-II; however, alphaKAP, beta2 CaMK-II and the RyR3 ryanodine receptor were also necessary for full CaMK-II activation. This is the first report to identify a direct Ca(2+)-sensitive target in left-right asymmetry and supports a model in which membrane targeted CaMK-II hetero-oligomers in nodal cells transduce the left-sided PKD2-dependent Ca(2+) signals to the LPM.


Subject(s)
Body Patterning , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Enzyme Activation , Epithelium/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Molecular Sequence Data , Sequence Alignment , Somites/enzymology
10.
Cell Tissue Res ; 339(3): 543-50, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20066548

ABSTRACT

Because of the permeability of the chorion, sea bass embryos are exposed to seawater before hatching and hence require precocious osmoregulatory processes. Several studies of other species have demonstrated the existence of ion-transporting cells located on the yolk sac membrane of embryos. In these cells, called ionocytes, ion movements are controlled by a pool of transmembrane proteins. Among them, the Na(+)/K(+)-ATPase, an abundant driving enzyme, has been used to reveal the presence or absence of ionocytes. We have immunostained the Na(+)/K(+)-ATPase in sea-bass embryos and shown the presence of the first ionocytes on the yolk sac membrane at stage 12 somites and the occurrence of ionocytes at other sites before hatching. Ionocytes located on the first gill slits have been identified at stage 14 somites. Primitive enteric ionocytes have also been detected at stage 14 somites in the mid and posterior gut. The presence of these cells might be related to the early opening of the gut to perivitelline fluids, both anteriorly by the gill slits and posteriorly by the anus. The role of embryonic ionocytes in osmoregulation before hatching is discussed.


Subject(s)
Bass/embryology , Embryo, Nonmammalian/cytology , Animals , Embryo, Nonmammalian/enzymology , Fertilization , Immunohistochemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Somites/cytology , Somites/enzymology
11.
Dev Dyn ; 238(11): 2867-76, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19795516

ABSTRACT

The process of segmentation in vertebrates is described by a clock and wavefront model consisting of a Notch signal and an fibroblast growth factor-8 (FGF8) gradient, respectively. To further investigate the segmentation process, we screened gene expression profiles for downstream targets of the segmentation clock. The Rnd1 and Rnd3 GTP-binding proteins comprise a subgroup of the Rho GTPase family that show a specific expression pattern similar to the Notch signal component ESR5, suggesting an association between Rnd1/3 and the segmentation clock. Rnd1/3 expression patterns are disrupted by overexpression of dominant-negative or active forms of Notch signaling genes, and responds to the FGF inhibitor SU5402 by a posterior shift analogous to other segmentation-related genes, suggesting that Rnd1/3 expressions are regulated by the segmentation clock machinery. We also show that antisense morpholino oligonucleotides to Rnd1/3 inhibit somite segmentation and differentiation in Xenopus embryos. These results suggest that Rnd1/3 are required for Xenopus somitogenesis.


Subject(s)
Embryo, Nonmammalian/embryology , GTP-Binding Proteins/metabolism , Receptors, Notch/metabolism , Somites/embryology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Embryo, Nonmammalian/enzymology , Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/metabolism , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Pyrroles/pharmacology , Somites/enzymology , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/metabolism , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics
12.
Mech Dev ; 126(3-4): 240-55, 2009.
Article in English | MEDLINE | ID: mdl-19027850

ABSTRACT

ADAM19 is a member of the meltrin subfamily of ADAM metalloproteases. In Xenopus, ADAM19 is present as a maternal transcript. Zygotic expression starts during gastrulation and is apparent in the dorsal blastopore lip. ADAM19 expression through neurulation and tailbud formation becomes enriched in dorsal structures such as the neural tube, the notochord and the somites. Using morpholino knock-down, we show that a reduction of ADAM19 protein in gastrula stage embryos results in a decrease of Brachyury expression in the notochord concomitant with an increase in the dorsal markers, Goosecoid and Chordin. These changes in gene expression are accompanied by a decrease in phosphorylated AKT, a downstream target of the EGF signaling pathway, and occur while the blastopore closes at the same rate as the control embryos. During neurulation and tailbud formation, ADAM19 knock-down induces a reduction of the neural markers N-tubulin and NRP1 but not Sox2. In the somitic mesoderm, the expression of MLC is also decreased while MyoD is not. ADAM19 knockdown also reduces neural crest markers prior to cell migration. Neural crest induction is also decreased in embryos treated with an EGF receptor inhibitor suggesting that this pathway is necessary for neural crest cell induction. Using targeted knock-down of ADAM19 we show that the reduction of neural and neural crest markers is cell autonomous and that the migration if the cranial neural crest is perturbed. We further show that ADAM19 protein reduction affects somite organization, reduces 12-101 expression and perturbs fibronectin localization at the intersomitic boundary.


Subject(s)
Muscle Development , Nervous System/embryology , Nervous System/enzymology , Neural Crest/embryology , Neural Crest/enzymology , Xenopus laevis/embryology , ADAM Proteins/genetics , ADAM Proteins/metabolism , Animals , Biomarkers/metabolism , Body Patterning/drug effects , Epidermal Growth Factor/metabolism , Fetal Proteins/metabolism , Gastrula/drug effects , Gastrula/embryology , Gastrula/enzymology , Gene Expression Regulation, Developmental/drug effects , Mesoderm/drug effects , Mesoderm/embryology , Muscle Development/drug effects , Nervous System/drug effects , Neural Crest/cytology , Neural Crest/drug effects , Neural Tube/drug effects , Neural Tube/embryology , Neural Tube/enzymology , Notochord/drug effects , Notochord/metabolism , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Somites/drug effects , Somites/embryology , Somites/enzymology , T-Box Domain Proteins/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Zygote/drug effects , Zygote/enzymology
13.
Dev Dyn ; 236(6): 1495-508, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17497689

ABSTRACT

The establishment of thresholds along morphogen gradients in the embryo is poorly understood. Using mathematical modeling, we show that mutually inhibitory gradients can generate and position sharp morphogen thresholds in the embryonic space. Taking vertebrate segmentation as a paradigm, we demonstrate that the antagonistic gradients of retinoic acid (RA) and Fibroblast Growth Factor (FGF) along the presomitic mesoderm (PSM) may lead to the coexistence of two stable steady states. Here, we propose that this bistability is associated with abrupt switches in the levels of FGF and RA signaling, which permit the synchronized activation of segmentation genes, such as mesp2, in successive cohorts of PSM cells in response to the segmentation clock, thereby defining the future segments. Bistability resulting from mutual inhibition of RA and FGF provides a molecular mechanism for the all-or-none transitions assumed in the "clock and wavefront" somitogenesis model. Given that mutually antagonistic signaling gradients are common in development, such bistable switches could represent an important principle underlying embryonic patterning.


Subject(s)
Fibroblast Growth Factors/antagonists & inhibitors , Fibroblast Growth Factors/metabolism , Signal Transduction , Tretinoin/antagonists & inhibitors , Tretinoin/metabolism , Animals , Body Patterning , Computer Simulation , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental , Humans , Kinetics , Models, Biological , Somites/enzymology , Somites/metabolism
14.
Dev Dyn ; 236(6): 1484-94, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17477400

ABSTRACT

Mesp2 and Paraxis are basic helix-loop-helix (bHLH) -type transcription factors coexpressed in the presomitic mesoderm (PSM) and are required for normal somite formation. Here, we show that Mesp2/Paraxis double-null mice exhibit a distinct phenotype unexpected from either Mesp2 or Paraxis single-null mice. In the posterior region of the body, most of the skeletal components of both the vertebral body and neural arches are severely reduced and only a rudimental lamina and ribs remain, indicating a strong genetic interaction in the sclerotomal cell lineage. However, yeast two-hybrid analyses revealed no direct interaction between Mesp2 and Paraxis. The Mesp2/Paraxis double-null embryo has caudalized somites, revealed by expanded Uncx4.1 expression pattern observed in the Mesp2-null embryo, but the expression level of Uncx4.1 was significantly decreased in mature somites, indicative of hypoplasia of lateral sclerotome derivatives. By focusing on vertebral column formation, we found that expressions of Pax1, Nkx3.1, and Bapx1 are regulated by Paraxis and that Pax9 expression was severely affected in the Mesp2/Paraxis double-null embryo. Furthermore, the expression of Pax3, a crucial factor for hypaxial muscle differentiation, is regulated by both Mesp2 and Paraxis in the anteriormost PSM and nascent somite region. The present data strongly suggest that patterning events by bHLH-type transcription factors have deep impacts on regional chondrogenic and myogenic differentiation of somitic cells, mainly by means of control of Pax genes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Polarity , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , PAX3 Transcription Factor , PAX9 Transcription Factor , Paired Box Transcription Factors/genetics , Protein Binding , Somites/cytology , Somites/enzymology , Somites/metabolism
15.
Dev Biol ; 305(2): 421-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17425953

ABSTRACT

Regulation of VEGFR-2 (Quek1) is an important mechanism during blood vessel formation. In the paraxial mesoderm, Quek1 expression is restricted to the lateral portion of the somite and later to sclerotomal cells surrounding the neural tube. By implanting FGF 8b/8c or SU 5402 beads into the paraxial mesoderm, we show that FGF8 in addition to BMP4 from the intermediate mesoderm (IM) is a positive regulator of VEGFR-2 (Quek1) expression in the quail embryo. The expression of Quek1 in the medial somite half is normally repressed by the notochord and Sfrps-expression in the neural tube. Over-expression of Wnt 1/3a also results in an up-regulation of Quek1 expression in the somites. We also show that up-regulation of FGF8/Wnt 1/3a leads to an increase in the number of endothelial cells, whereas inhibition of FGF and Wnt signaling by SU 5402 and Sfrp-2 results in a loss of endothelial cells. Our results demonstrate that the regulation of Quek1 expression in the somites is mediated by the cooperative actions of BMP4, FGF8 and Wnt-signaling pathways.


Subject(s)
Bone Morphogenetic Proteins/physiology , Coturnix/embryology , Fibroblast Growth Factor 8/physiology , Receptors, Neurotransmitter/biosynthesis , Somites/enzymology , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Wnt Proteins/physiology , Wnt1 Protein/physiology , Animals , Avian Proteins/biosynthesis , Avian Proteins/genetics , Avian Proteins/physiology , Bone Morphogenetic Protein 4 , Cells, Cultured , Coturnix/metabolism , Endothelial Cells/enzymology , Enzyme Induction/genetics , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Mice , Receptors, Neurotransmitter/genetics , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/genetics , Wnt3 Protein
16.
Dev Dyn ; 235(12): 3327-35, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17061267

ABSTRACT

A very dynamic and localised spatiotemporal expression pattern of Sulf1 was observed in axial structures and different regions of developing quail somites that included myotomal and sclerotomal regions at specific levels. Sulf1 expression was also observed in not only the scapular and pelvic girdle forming regions of the quail limb that connect the appendicular skeleton to the body trunk but also the cartilage templates of the appendicular skeleton. The highest expression level of Sulf1 was observed in condensing mesenchyme, during the early differentiation stage of chondrogenesis, and highly dynamic expression was observed in the perichondrial and joint-forming regions. Overexpression of Sulf1 in quail micromass cultures enhanced aggregation and differentiation of prechondrocytes into chondrogenic lineage supporting its role in mesenchymal condensation and early differentiation of cartilaginous elements. The exposure of digital explants to high levels of Sulf1 expression in vitro led to increased growth of the original 1st phalange but complete inhibition of joint formation and generation of any further phalanges. Sulf1 thus plays a key role during multiple stages of cartilage development and joint formation.


Subject(s)
Quail/embryology , Quail/genetics , Sulfotransferases/genetics , Animals , Cartilage/embryology , Cartilage/enzymology , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/enzymology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Extremities/embryology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , In Situ Hybridization , Joints/embryology , Joints/enzymology , Mesoderm/enzymology , Mutation , Quail/metabolism , Signal Transduction , Somites/enzymology
17.
Dev Dyn ; 235(10): 2881-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16921498

ABSTRACT

The protein related to Dan and Cerberus, or PRDC, is a secreted glycoprotein, which belongs to the DAN subfamily of bone morphogenetic protein (BMP) antagonists. In zebrafish, prdc is expressed initially around 17 hours postfertilization in the developing eyes and the first two pharyngeal arches. Expression in the eye starts in the outer layers of the optic cup. Later, prdc expression domains are juxtaposed at the edges of the optic cup surrounding the choroid fissure, then gradually becoming restricted to a small site in the ventral marginal zone. Prdc expression in the arch mesenchyme expands stepwise to the remaining posterior arches. Prdc is also detectable in the ventral part of the somites and the mesenchyme of the swim bladder. The relatively late appearance during development is a unique feature of Prdc among BMP antagonists. Moreover, the complexity of the prdc expression pattern suggests possible roles in eye development, pharyngeal arch remodeling, somitogenesis, and swim bladder organogenesis.


Subject(s)
Air Sacs/metabolism , Branchial Region/metabolism , Eye/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Somites/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Air Sacs/embryology , Amino Acid Sequence , Animals , Branchial Region/embryology , Eye/embryology , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Somites/enzymology , Zebrafish/embryology
18.
Dev Biol ; 288(1): 73-86, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16248994

ABSTRACT

The p38 MAPK signaling pathway is essential for skeletal muscle differentiation in tissue culture models. We demonstrate a novel role for p38 MAPK in myogenesis during early Xenopus laevis development. Interfering with p38 MAPK causes distinct defects in myogenesis. The initial expression of Myf5 is selectively blocked, while expression of MyoD is unaffected. Expression of a subset of muscle structural genes is reduced. Convergent extension movements are prevented and segmentation of the paraxial mesoderm is delayed, probably due to the failure of cells to withdraw from the cell cycle. Myotubes are properly formed; however, at later stages, they begin to degenerate, and the boundaries between somites disappear. Significant apoptotic cell death occurs in most parts of the somites. The ventral body wall muscle derived from migratory progenitor cells of the ventral somite region is poorly formed. Our data indicate that the developmental defects caused by p38alpha-knockdown were mediated by the loss of XMyf5 expression. Thus, this study identifies a specific intracellular pathway in which p38 MAPK and Myf5 proteins regulate a distinct myogenic program.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal/embryology , Muscle, Skeletal/enzymology , Myogenic Regulatory Factor 5/biosynthesis , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Apoptosis/physiology , Body Patterning/physiology , Cell Cycle/physiology , Muscle, Skeletal/cytology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenic Regulatory Factor 5/genetics , Phenotype , Somites/cytology , Somites/enzymology , Somites/physiology , Xenopus laevis
19.
Dev Biol ; 286(2): 405-14, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16112664

ABSTRACT

Remodeling of the extracellular matrix (ECM) during development, angiogenesis, wound healing, tumor metastasis, and other morphogenetic processes depends on the exquisitely regulated activities of matrix metalloproteinases (MMPs). Yet very little is known about the activity patterns of these proteases in vivo. We have employed fluorescent MMP-substrates, both in vitro and in vivo, to characterize patterns of MMP activity in the zebrafish embryo. Qualitatively similar patterns of degradation are detected using native Type I or Type IV collagen substrates, suggesting that multiple MMPs are being regulated concomitantly. MMP activity is observed primarily in ECM-rich structures predicted to be undergoing active remodeling, such as the perichordal sheath and somite boundaries. Patterns of Type I and Type IV collagen hydrolysis are similar, but not identical in embryos of any given stage. Conventional gelatin zymography shows MMPs present in embryos as early as 3-somites (11 h) and our in vivo assays detect Type IV collagen degradation at somite boundaries as early as 4-somites (11.5 h). However, we are unable to detect significant in vitro activity using homogenates made from embryos prior to Prim-16 (31 h). Mixed lysate assays demonstrate that this is the result of endogenous inhibitors present in early embryos, suggesting a model of matrix remodeling regulated by spatially heterogeneous MMP inhibition.


Subject(s)
Embryo, Nonmammalian/enzymology , Matrix Metalloproteinases/metabolism , Zebrafish/embryology , Animals , Collagen Type I/metabolism , Collagen Type IV/metabolism , Electrophoresis, Agar Gel , Extracellular Matrix/metabolism , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/analysis , Morphogenesis , Protease Inhibitors , Somites/enzymology , Somites/metabolism
20.
Development ; 132(6): 1305-14, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15716340

ABSTRACT

Cells in the early vertebrate somite receive cues from surrounding tissues, which are important for their specification. A number of signalling pathways involved in somite patterning have been described extensively. By contrast, the interactions between cells from different regions within the somite are less well characterised. Here, we demonstrate that myotomally derived FGFs act through the MAPK signal transduction cascade and in particular, ERK1/2 to activate scleraxis expression in a population of mesenchymal progenitor cells in the dorsal sclerotome. We show that the levels of active, phosphorylated ERK protein in the developing somite are crucial for the expression of scleraxis and Mkp3. MKP3 is a dual specificity phosphatase and a specific antagonist of ERK MAP kinases and we demonstrate that in somites Mkp3 transcription depends on the presence of active ERK. Therefore, MKP3 and ERK MAP kinase constitute a negative feedback loop activated by FGF in sclerotomal progenitor cells. We propose that tight control of ERK signalling strength by MKP3 is important for the appropriate regulation of downstream cellular responses including the activation of scleraxis. We show that increased or decreased levels of phosphorylated ERK result in the loss of scleraxis transcripts and the loss of distal rib development, highlighting the importance of the MKP3-ERK-MAP kinase mediated feedback loop for cell specification and differentiation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Protein Tyrosine Phosphatases/metabolism , Ribs/embryology , Somites/enzymology , Transcription Factors/genetics , Animals , Avian Proteins , Basic Helix-Loop-Helix Transcription Factors , Chick Embryo , Dual Specificity Phosphatase 6 , Fibroblast Growth Factors/metabolism , RNA, Messenger/metabolism , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...