Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
J Dent Res ; 98(1): 98-106, 2019 01.
Article in English | MEDLINE | ID: mdl-30325689

ABSTRACT

Crown shapes in mammalian teeth vary considerably from species to species, and morphological characters in crown shape have been used to identify species. Cusp pattern is one of the characters in crown shape. In the processes governing the formation of cusp pattern, the Shh pathway has been implicated as an important player. Suppression of Shh signaling activity in vitro in explant assays appears to induce supernumerary cusp formation in wild-type tooth germs. However, the in vivo role of Shh signaling in cusp pattern formation and the molecular mechanisms by which Shh regulates cusp patterning are not clear. Here, through in vivo phenotypic analyses of mice in which Shh activity was suppressed and compared with wild-type mice, we characterized differences in the location, number, incidence, and shape of supernumerary cusps in molars at embryonic day 15.5. We found that the distances between cusps were reduced in molars of Shh activity-suppressed mice in vivo. These findings confirm and extend the previous idea that Shh acts as an inhibitor in the reaction-diffusion model for cusp pattern formation by negatively regulating the intercuspal distance. We uncovered a significant reduction of expression level of Sostdc1, which encodes a secreted modulator of Wnt signaling, after suppression of Shh activity. The supernumerary cusp formation in Sostdc1-/- mice and compound Sostdc1 and Lrp mutant mice indicates a strong association between Wnt and Shh signaling pathways in cusp patterning. In further support of this idea, there is a high degree of similarity in the supernumerary cusp patterns of mice lacking Sostdc1 or Shh at embryonic day 15.5. These results suggest that Shh plays an inhibitory role in cusp pattern formation by modulating Wnt signaling through the positive regulation of Sostdc1.


Subject(s)
Body Patterning/genetics , Bone Morphogenetic Proteins/physiology , Hedgehog Proteins/physiology , Tooth/embryology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Body Patterning/physiology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Molar , Signal Transduction , Tooth/metabolism , Tooth Crown , Tooth Germ , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway
2.
J Dent Res ; 96(11): 1306-1313, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28732181

ABSTRACT

Rare mutations in IRF6 and GRHL3 cause Van der Woude syndrome, an autosomal dominant orofacial clefting disorder. Common variants in IRF6 and GRHL3 also contribute risk for isolated orofacial clefting. Similarly, variants within genes that encode receptor tyrosine kinase (RTK) signaling components, including members of the FGF pathway, EPHA3 and SPRY2, also contribute risk for isolated orofacial clefting. In the mouse, loss of Irf6 or perturbation of Fgf signaling leads to abnormal oral epithelial adhesions and cleft palate. Oral adhesions can result from a disruption of periderm formation. Here, we find that IRF6 and SPRY4 signaling interact in periderm function. We crossed Irf6 heterozygous ( Irf6+/-) mice with transgenic mice that express Spry4 in the basal epithelial layer ( TgKRT14::Spry4). While embryos with either of these mutations can have abnormal oral adhesions, using a new quantitative assay, we observed a nonadditive effect of abnormal oral epithelial adhesions in the most severely affected double mutant embryos ( Irf6+/-;TgKRT14::Spry4). At the molecular level, the sites of abnormal oral adhesions maintained periderm-like cells that express keratin 6, but we observed abnormal expression of GRHL3. Together, these data suggest that Irf6 and RTK signaling interact in regulating periderm differentiation and function, as well as provide a rationale to screen for epistatic interactions between variants in IRF6 and RTK signaling pathway genes in human orofacial clefting populations.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Interferon Regulatory Factors/genetics , Nerve Tissue Proteins/genetics , Tissue Adhesions/genetics , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Animals , Cleft Lip/embryology , Cleft Palate/embryology , Cysts/embryology , Cysts/genetics , Disease Models, Animal , Jaw Abnormalities/embryology , Jaw Abnormalities/genetics , Lip/abnormalities , Lip/embryology , Mice , Mice, Transgenic , Mouth Abnormalities/embryology , Mouth Abnormalities/genetics , Mutation , Phenotype , Signal Transduction , Tissue Adhesions/embryology
3.
J Dent Res ; 96(11): 1273-1281, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28692808

ABSTRACT

Cleft palate is a common birth defect caused by disruption of palatogenesis during embryonic development. Although mutations disrupting components of the Wnt signaling pathway have been associated with cleft lip and palate in humans and mice, the mechanisms involving canonical Wnt signaling and its regulation in secondary palate development are not well understood. Here, we report that canonical Wnt signaling plays an important role in Pax9-mediated regulation of secondary palate development. We found that cleft palate pathogenesis in Pax9-deficient embryos is accompanied by significantly reduced expression of Axin2, an endogenous target of canonical Wnt signaling, in the developing palatal mesenchyme, particularly in the posterior regions of the palatal shelves. We found that expression of Dkk2, encoding a secreted Wnt antagonist, is significantly increased whereas the levels of active ß-catenin protein, the essential transcriptional coactivator of canonical Wnt signaling, is significantly decreased in the posterior regions of the palatal shelves in embryonic day 13.5 Pax9-deficent embryos in comparison with control littermates. We show that small molecule-mediated inhibition of Dickkopf (DKK) activity in utero during palatal shelf morphogenesis partly rescued secondary palate development in Pax9-deficient embryos. Moreover, we found that genetic inactivation of Wise, which is expressed in the developing palatal shelves and encodes another secreted antagonist of canonical Wnt signaling, also rescued palate morphogenesis in Pax9-deficient mice. Furthermore, whereas Pax9del/del embryos exhibit defects in palatal shelf elevation/reorientation and significant reduction in accumulation of hyaluronic acid-a high molecular extracellular matrix glycosaminoglycan implicated in playing an important role in palatal shelf elevation-80% of Pax9del/del;Wise-/- double-mutant mouse embryos exhibit rescued palatal shelf elevation/reorientation, accompanied by restored hyaluronic acid accumulation in the palatal mesenchyme. Together, these data identify a crucial role for canonical Wnt signaling in acting downstream of Pax9 to regulate palate morphogenesis.


Subject(s)
Cleft Palate/embryology , Cleft Palate/genetics , Paired Box Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Embryonic Development , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Mice , Morphogenesis , PAX9 Transcription Factor , Palate/embryology , Signal Transduction/genetics , Transcription Factors/genetics , Wnt Proteins/genetics , beta Catenin/genetics
4.
Bone ; 76: 58-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25836764

ABSTRACT

The response of the skeleton to loading appears to be mediated through the activation of the Wnt/ß-catenin signaling pathway and osteocytes have long been postulated to be the primary mechanosensory cells in bone. To examine the kinetics of the mechanoresponse of bone and cell types involved in vivo, we performed forearm loading of 17-week-old female TOPGAL mice. ß-catenin signaling was observed only in embedded osteocytes, not osteoblasts, at 1h post-loading, spreading to additional osteocytes and finally to cells on the bone surface by 24h. This early activation at 1h appeared to be independent of receptor (Lrp5/6) mediated activation as it occurred in the presence of the inhibitors sclerostin and/or Dkk1. The COX-2 inhibitor, Carprofen, blocked the activation of ß-catenin signaling and decline in sclerostin positive osteocytes post-loading implying an important role for prostaglandin. In vitro, PI3K/Akt activation was shown to be required for ß-catenin nuclear translocation downstream from prostaglandin in MLO-Y4 osteocyte-like cells supporting this mechanism. Downstream targets of ß-catenin signaling, sclerostin and Dkk1, were also examined and found to be significantly downregulated in osteocytes in vivo at 24h post-loading. The pattern of initially activated osteocytes appeared random and in order to understand this heterogeneous expression, a novel finite element model of the strain field in the ulna was developed, which predicts highly variable local magnitudes of strain experienced by osteocytes. In summary, both in vivo and in vitro models show the rapid activation of ß-catenin in response to load through the early release of prostaglandin and that strain fields in the bone are extremely heterogeneous resulting in heterogeneous activation of the ß-catenin pathway in osteocytes in vivo.


Subject(s)
Osteocytes/metabolism , Prostaglandins/metabolism , Signal Transduction , Stress, Mechanical , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Female , Finite Element Analysis , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kinetics , Mice , beta Catenin/genetics
7.
Curr Opin Cell Biol ; 13(6): 698-705, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11698185

ABSTRACT

Proper craniofacial development requires the orchestrated integration of multiple specialized tissue interactions. Recent analyses suggest that craniofacial development is not dependent upon neural crest pre-programming as previously thought but is regulated by a more complex integration of cell and tissue interactions. In the absence of neural crest cells it is still possible to obtain normal arch patterning indicating that neural crest is not responsible for patterning all of arch development. The mesoderm, endoderm and surface ectoderm tissues play a role in the patterning of the branchial arches, and there is now strong evidence that Hoxa2 acts as a selector gene for the pathways that govern second arch structures.


Subject(s)
Body Patterning , Branchial Region/embryology , Homeodomain Proteins/physiology , Neural Crest/physiology , Animals , Bone and Bones/embryology , Ectoderm/physiology , Endoderm/physiology , Gene Expression Regulation, Developmental , Head/embryology , Homeodomain Proteins/genetics , Mesoderm/physiology , Models, Biological , Neural Crest/cytology , Rhombencephalon/embryology , Rhombencephalon/physiology
8.
Development ; 128(15): 3017-27, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11532923

ABSTRACT

Hoxa1 and Hoxb1 have overlapping synergistic roles in patterning the hindbrain and cranial neural crest cells. The combination of an ectoderm-specific regulatory mutation in the Hoxb1 locus and the Hoxa1 mutant genetic background results in an ectoderm-specific double mutation, leaving the other germ layers impaired only in Hoxa1 function. This has allowed us to examine neural crest and arch patterning defects that originate exclusively from the neuroepithelium as a result of the simultaneous loss of Hoxa1 and Hoxb1 in this tissue. Using molecular and lineage analysis in this double mutant background we demonstrate that presumptive rhombomere 4, the major site of origin of the second pharyngeal arch neural crest, is reduced in size and has lost the ability to generate neural crest cells. Grafting experiments using wild-type cells in cultured normal or double mutant mouse embryos demonstrate that this is a cell-autonomous defect, suggesting that the formation or generation of cranial neural crest has been uncoupled from segmental identity in these mutants. Furthermore, we show that loss of the second arch neural crest population does not have any adverse consequences on early patterning of the second arch. Signalling molecules are expressed correctly and pharyngeal pouch and epibranchial placode formation are unaffected. There are no signs of excessive cell death or loss of proliferation in the epithelium of the second arch, suggesting that the neural crest cells are not the source of any indispensable mitogenic or survival signals. These results illustrate that Hox genes are not only necessary for proper axial specification of the neural crest but that they also play a vital role in the generation of this population itself. Furthermore, they demonstrate that early patterning of the separate components of the pharyngeal arches can proceed independently of neural crest cell migration.


Subject(s)
Body Patterning , Branchial Region/embryology , Homeodomain Proteins/physiology , Neural Crest/embryology , Transcription Factors/physiology , Animals , Cell Lineage , Cell Movement , Cell Survival , Culture Techniques , Homeodomain Proteins/genetics , Mice , Mutation , Neural Crest/cytology , Transcription Factors/genetics
9.
Development ; 128(18): 3595-607, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566863

ABSTRACT

During development of the vertebrate hindbrain, Hox genes play multiple roles in the segmental processes that regulate anteroposterior (AP) patterning. Paralogous Hox genes, such as Hoxa3, Hoxb3 and Hoxd3, generally have very similar patterns of expression, and gene targeting experiments have shown that members of paralogy group 3 can functionally compensate for each other. Hence, distinct functions for individual members of this family may primarily depend upon differences in their expression domains. The earliest domains of expression of the Hoxa3 and Hoxb3 genes in hindbrain rhombomeric (r) segments are transiently regulated by kreisler, a conserved Maf b-Zip protein, but the mechanisms that maintain expression in later stages are unknown. In this study, we have compared the segmental expression and regulation of Hoxa3 and Hoxb3 in mouse and chick embryos to investigate how they are controlled after initial activation. We found that the patterns of Hoxa3 and Hoxb3 expression in r5 and r6 in later stages during mouse and chick hindbrain development were differentially regulated. Hoxa3 expression was maintained in r5 and r6, while Hoxb3 was downregulated. Regulatory comparisons of cis-elements from the chick and mouse Hoxa3 locus in both transgenic mouse and chick embryos have identified a conserved enhancer that mediates the late phase of Hoxa3 expression through a conserved auto/cross-regulatory loop. This block of similarity is also present in the human and horn shark loci, and contains two bipartite Hox/Pbx-binding sites that are necessary for its in vivo activity in the hindbrain. These HOX/PBC sites are positioned near a conserved kreisler-binding site (KrA) that is involved in activating early expression in r5 and r6, but their activity is independent of kreisler. This work demonstrates that separate elements are involved in initiating and maintaining Hoxa3 expression during hindbrain segmentation, and that it is regulated in a manner different from Hoxb3 in later stages. Together, these findings add further strength to the emerging importance of positive auto- and cross-regulatory interactions between Hox genes as a general mechanism for maintaining their correct spatial patterns in the vertebrate nervous system.


Subject(s)
Avian Proteins , Body Patterning/genetics , Homeodomain Proteins/genetics , Oncogene Proteins , Rhombencephalon/embryology , Xenopus Proteins , Animals , Base Sequence , Binding Sites , Biological Evolution , Chick Embryo , Conserved Sequence , DNA-Binding Proteins , Enhancer Elements, Genetic , G-Box Binding Factors , Gene Expression Regulation, Developmental , MafB Transcription Factor , Mice , Mice, Transgenic , Models, Genetic , Regulatory Sequences, Nucleic Acid , Sequence Homology, Nucleic Acid , Species Specificity , Transcription Factors
11.
Dev Biol ; 233(2): 468-81, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11336508

ABSTRACT

During hindbrain development, segmental regulation of the paralogous Hoxa2 and Hoxb2 genes in rhombomeres (r) 3 and 5 involves Krox20-dependent enhancers that have been conserved during the duplication of the vertebrate Hox clusters from a common ancestor. Examining these evolutionarily related control regions could provide important insight into the degree to which the basic Krox20-dependent mechanisms, cis-regulatory components, and their organization have been conserved. Toward this goal we have performed a detailed functional analysis of a mouse Hoxa2 enhancer capable of directing reporter expression in r3 and r5. The combined activities of five separate cis-regions, in addition to the conserved Krox20 binding sites, are involved in mediating enhancer function. A CTTT (BoxA) motif adjacent to the Krox20 binding sites is important for r3/r5 activity. The BoxA motif is similar to one (Box1) found in the Hoxb2 enhancer and indicates that the close proximity of these Box motifs to Krox20 sites is a common feature of Krox20 targets in vivo. Two other rhombomeric elements (RE1 and RE3) are essential for r3/r5 activity and share common TCT motifs, indicating that they interact with a similar cofactor(s). TCT motifs are also found in the Hoxb2 enhancer, suggesting that they may be another common feature of Krox20-dependent control regions. The two remaining Hoxa2 cis-elements, RE2 and RE4, are not conserved in the Hoxb2 enhancer and define differences in some of components that can contribute to the Krox20-dependent activities of these enhancers. Furthermore, analysis of regulatory activities of these enhancers in a Krox20 mutant background has uncovered differences in their degree of dependence upon Krox20 for segmental expression. Together, this work has revealed a surprising degree of complexity in the number of cis-elements and regulatory components that contribute to segmental expression mediated by Krox20 and sheds light on the diversity and evolution of Krox20 target sites and Hox regulatory elements in vertebrates.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Rhombencephalon/embryology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Base Sequence , DNA/genetics , Early Growth Response Protein 2 , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Genes, Homeobox , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Molecular Sequence Data , Sequence Homology, Nucleic Acid
12.
Dev Biol ; 232(1): 176-90, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11254356

ABSTRACT

The expression pattern of the mouse Hoxb3 gene is exceptionally complex and dynamic compared with that of other members of the Hoxb cluster. There are multiple types of transcripts for Hoxb3 gene, and the anterior boundaries of its expression vary at different stages of development. Two enhancers flanking Hoxb3 on the 3' and 5' sides regulate Hoxb2 and Hoxb4, respectively, and these control regions define the two ends of a 28-kb interval in and around the Hoxb3 locus. To assay the regulatory potential of DNA fragments in this interval we have used transgenic analysis with a lacZ reporter gene to locate cis-elements for directing the dynamic patterns of Hoxb3 expression. Our detailed analysis has identified four new and widely spaced cis-acting regulatory regions that can together account for major aspects of the Hoxb3 expression pattern. Elements Ib, IIIa, and IVb control gene expression in neural and mesodermal tissues; element Va controls mesoderm-specific gene expression. The most anterior neural expression domain of Hoxb3 is controlled by an r5 enhancer (element IVa); element IIIa directs reporter expression in the anterior spinal cord and hindbrain up to r6, and the region A enhancer (in element I) mediates posterior neural expression. Hence, the regulation of segmental expression of Hoxb3 in the hindbrain is different from that of Hoxa3, as two separate enhancer elements contribute to expression in r5 and r6. The mesoderm-specific element (Va) directs reporter expression to prevertebra C1 at 12.5 dpc, which is the anterior limit of paraxial mesoderm expression for Hoxb3. When tested in combinations, these cis-elements appear to work as modules in an additive manner to recapitulate the major endogenous expression patterns of Hoxb3 during embryogenesis. Together our study shows that multiple control elements direct reporter gene expression in diverse tissue-, temporal-, and spatially restricted subset of the endogenous Hoxb3 expression domains and work in concert to control the neural and mesodermal patterns of expression.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Xenopus Proteins , Animals , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
13.
J Biol Chem ; 276(23): 20506-15, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11278854

ABSTRACT

Regionally restricted expression patterns of Hox genes in developing embryos rely on auto-, cross-, and para-regulatory transcriptional elements. One example is the Hoxb1 auto-regulatory element (b1-ARE), which drives expression of Hoxb1 in the fourth rhombomere of the hindbrain. We previously showed that HOXB1 and PBX1 activate transcription from the b1-ARE by binding to sequences required for the expression of a reporter gene in rhombomere 4 in vivo. We now report that in embryonal carcinoma cells, which retain characteristics of primitive neuroectodermal cells, the b1-ARE displays higher basal and HOX/PBX-induced activities than in other cell backgrounds. We have identified a bipartite-binding site for SOX/OCT heterodimers within the b1-ARE that accounts for its cell context-specific activity and is required for maximal transcriptional activity of HOX/PBX complexes in embryonal carcinoma cells. Furthermore, we found that in an embryonal carcinoma cell background, HOXB1 has a significantly higher transcriptional activity than its paralog HOXA1. We map the determinants for this differential activity within the HOXB1 N-terminal transcriptional activation domain. By using analysis in transgenic and HOXA1 mutant mice, we extended these findings on the differential activities of HOXA1 and HOXB1 in vivo, and we demonstrated that they are important for regulating aspects of HOXB1 expression in the hindbrain. We found that mutation of the SOX/OCT site and targeted inactivation of Hoxa1 both impair the response of the b1-ARE to retinoic acid in transgenic mice. Our results show that Hoxa1 is the primary mediator of the response of b1-ARE to retinoic acid in vivo and that this function is dependent on the binding of SOX/OCT heterodimers to the b1-ARE. These results uncover novel functional differences between Hox paralogs and their modulators.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , DNA Probes , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Transcription Factors/physiology , Transcriptional Activation/physiology , Tretinoin/pharmacology
14.
Mech Dev ; 100(1): 141-4, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11118901

ABSTRACT

Defects in the Notch pathway ligand Dll3 have been identified in the mouse pudgy (Dll3(pu)) and human spondylocostal dysostosis (SD, MIM 277300) mutations. Although these mutations are primarily associated with segmental defects in the axial skeleton and somitic patterning, they also exhibit cranial neurological defects. Therefore we have looked at the expression of Dll3 in the developing mouse nervous system. The expression of Notch ligands and receptors shares common features at 10.75 dpc in the rhombic lips and dorsal hindbrain. Temporal analysis of Dll3 expression from 9.0 to 11.0 dpc reveals that it is strongly expressed in laminar columns linked with regions of neuronal differentiation and hindbrain segmentation. Transverse sections show that Dll3 is expressed in territories where commissural neurons are formed. We have also looked at neuronal patterning in the mid-hindbrain region in Dll3(pu) mutants.


Subject(s)
Membrane Proteins/genetics , Nervous System/embryology , Transcription Factors , Animals , Brain/metabolism , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Membrane Proteins/biosynthesis , Mice , Neurons/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor, Notch1 , Receptor, Notch2 , Receptor, Notch4 , Receptors, Cell Surface/biosynthesis , Receptors, Notch , Rhombencephalon/embryology , Time Factors
15.
Dev Biol ; 239(1): 148-60, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11784025

ABSTRACT

In order to identify factors involved in posteriorization of the central nervous system, we undertook a functional screen in Xenopus animal cap explants which involved coinjecting noggin RNA together with pools of RNA from a chick somite cDNA library. In the course of this screen, we isolated a clone encoding a truncated form of beta-catenin, which induced posterior neural and dorsal mesodermal markers when coinjected with noggin in animal caps. Similar results were obtained with Xwnt-8 and Xwnt-3a, suggesting that these effects are a consequence of activating the canonical Wnt signalling pathway. To investigate whether the activation of posterior neural markers requires mesoderm induction, we performed experiments using a chimeric inducible form of beta-catenin. Activation of this protein during blastula stages resulted in the induction of both posterior neural and mesodermal markers, while activation during gastrula stages induced only posterior neural markers. We show that this posteriorizing activity occurs by an indirect and noncell-autonomous mechanism requiring FGF signalling.


Subject(s)
Body Patterning , Cytoskeletal Proteins/metabolism , Fibroblast Growth Factors/metabolism , Nervous System/embryology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators , Xenopus/embryology , Zebrafish Proteins , Animals , Biomarkers/analysis , Blotting, Western , Carrier Proteins , Cytoskeletal Proteins/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryonic Induction , In Situ Hybridization , Mesoderm/metabolism , Nervous System/cytology , Nervous System/metabolism , Neurons/metabolism , Proteins/analysis , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion/genetics , Wnt Proteins , Xenopus/metabolism , Xenopus Proteins , beta Catenin
16.
Nature ; 408(6814): 854-7, 2000 Dec 14.
Article in English | MEDLINE | ID: mdl-11130723

ABSTRACT

The comparison of Hox genes between vertebrates and their closest invertebrate relatives (amphioxus and ascidia) highlights two derived features of Hox genes in vertebrates: duplication of the Hox gene cluster, and an elaboration of Hox expression patterns and roles compared with non-vertebrate chordates. We have investigated how new expression domains and their associated developmental functions evolved, by testing the cis-regulatory activity of genomic DNA fragments from the cephalochordate amphioxus Hox cluster in transgenic mouse and chick embryos. Here we present evidence for the conservation of cis-regulatory mechanisms controlling gene expression in the neural tube for half a billion years of evolution, including a dependence on retinoic acid signalling. We also identify amphioxus Hox gene regulatory elements that drive spatially localized expression in vertebrate neural crest cells, in derivatives of neurogenic placodes and in branchial arches, despite the fact that cephalochordates lack both neural crest and neurogenic placodes. This implies an elaboration of cis-regulatory elements in the Hox gene cluster of vertebrate ancestors during the evolution of craniofacial patterning.


Subject(s)
Biological Evolution , Chordata, Nonvertebrate/embryology , Gene Expression Regulation, Developmental , Genes, Homeobox , Head , Vertebrates/embryology , Animals , Animals, Genetically Modified , Body Patterning , Central Nervous System/embryology , Chick Embryo , Conserved Sequence , Culture Techniques , Evolution, Molecular , Head/embryology , Mice , Multigene Family , Neural Crest/embryology , Regulatory Sequences, Nucleic Acid , Signal Transduction , Tretinoin/physiology
17.
Curr Opin Genet Dev ; 10(4): 380-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10889064

ABSTRACT

Retinoid signalling has been implicated in regulating a wide variety of processes in vertebrate development. Recent advances from analyses on the synthesis, degradation and distribution of retinoids in combination with functional analysis of signalling components have provided important insights into the regulation of patterning the nervous system and the hindbrain in particular.


Subject(s)
Body Patterning/physiology , Rhombencephalon/embryology , Signal Transduction/physiology , Tretinoin/metabolism , Animals , Humans , Morphogenesis , Retinoids/metabolism
19.
Mech Dev ; 94(1-2): 147-56, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10842066

ABSTRACT

dreher is a spontaneous mouse mutation in which adult animals display a complex phenotype associated with hearing loss, neurological, pigmentation and skeletal abnormalities. During early embryogenesis, the neural tube of dreher mutants is abnormally shaped in the region of the rhomboencephalon, due to problems in the formation of a proper roof plate over the otic hindbrain. We have studied the expression of Hox/lacZ transgenic mouse strains in the dreher background and shown that primary segmentation of the neural tube is not altered in these mutants, although correct morphogenesis is affected resulting in misshapen rhombomeres. Neural crest derivatives from rhombomere 6, such as the glossopharyngeal ganglion, are defective, and the dorsal neural tube marker Wnt1 is absent from this segment. Selected trunk neural crest populations are also altered, as there is a lack of pigmentation in the thoracic region of mutant mice. Skeletal defects include abnormal cranial bones of neural crest origin, and improper fusion of the dorsal aspects of cervical and thoracic vertebrae. Taken together, the gene affected in the dreher mutant is responsible for correct patterning of the dorsal-most cell types of the neural tube, that is, the neural crest and the roof plate, in the hindbrain region. Axial skeletal defects could reflect inductive influence of the dorsal neural tube on proper fusion of the neural arches. It is possible that a common precursor population for both neural crest and roof plate is the cellular target of the dreher mutation.


Subject(s)
Body Patterning/genetics , Central Nervous System/embryology , Mutation , Rhombencephalon/embryology , Skeleton , Zebrafish Proteins , Animals , Bone and Bones/abnormalities , Central Nervous System/growth & development , Female , Gene Expression Regulation, Developmental , Genes, Homeobox , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Motor Neurons/physiology , Neural Crest/embryology , Neurons, Afferent/physiology , Proto-Oncogene Proteins/metabolism , Rhombencephalon/growth & development , Wnt Proteins , Wnt1 Protein
20.
Nat Genet ; 24(4): 438-41, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742114

ABSTRACT

Spondylocostal dysostosis (SD, MIM 277300) is a group of vertebral malsegmentation syndromes with reduced stature resulting from axial skeletal defects. SD is characterized by multiple hemivertebrae, rib fusions and deletions with a non-progressive kyphoscoliosis. Cases may be sporadic or familial, with both autosomal dominant and autosomal recessive modes of inheritance reported. Autosomal recessive SD maps to a 7.8-cM interval on chromosome 19q13.1-q13.3 that is homologous with a mouse region containing a gene encoding the Notch ligand delta-like 3 (Dll3). Dll3 is mutated in the X-ray-induced mouse mutant pudgy (pu), causing a variety of vertebrocostal defects similar to SD phenotypes. Here we have cloned and sequenced human DLL3 to evaluate it as a candidate gene for SD and identified mutations in three autosomal recessive SD families. Two of the mutations predict truncations within conserved extracellular domains. The third is a missense mutation in a highly conserved glycine residue of the fifth epidermal growth factor (EGF) repeat, which has revealed an important functional role for this domain. These represent the first mutations in a human Delta homologue, thus highlighting the critical role of the Notch signalling pathway and its components in patterning the mammalian axial


Subject(s)
Dysostoses/genetics , Membrane Proteins/genetics , Ribs/abnormalities , Scoliosis/genetics , Spine/abnormalities , Adult , Animals , Child , Chromosomes, Human, Pair 19/genetics , Cloning, Molecular , Conserved Sequence , DNA Mutational Analysis , Dysostoses/diagnostic imaging , Dysostoses/etiology , Female , Genetic Linkage , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Mice , Molecular Sequence Data , Mutation , Pedigree , Protein Structure, Tertiary/genetics , Radiography , Receptors, Notch , Ribs/diagnostic imaging , Scoliosis/diagnostic imaging , Scoliosis/etiology , Sequence Homology, Amino Acid , Signal Transduction/genetics , Spine/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...