Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters










Publication year range
1.
Oncogene ; 28(27): 2513-23, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19448672

ABSTRACT

Inappropriate kinase expression and subsequent promiscuous activity defines the transformation of many solid tumors including renal cell carcinoma (RCC). Thus, the expression of novel tumor-associated kinases has the potential to dramatically shape tumor cell behavior. Further, identifying tumor-associated kinases can lend insight into patterns of tumor growth and characteristics. Here, we report the identification of the RTK-like orphan receptor 2 (Ror2), a new tumor-associated kinase in RCC cell lines and primary tumors. Ror2 is an orphan receptor tyrosine kinase with physiological expression normally seen in the embryonic kidney. However, in RCC, Ror2 expression correlated with expression of genes involved at the extracellular matrix, including Twist and matrix metalloprotease-2 (MMP2). Expression of MMP2 in RCC cells was suppressed by Ror2 knockdown, placing Ror2 as a mediator of MMP2 regulation in RCC and a potential regulator of extracellular matrix remodeling. The suppression of Ror2 not only inhibited cell migration, but also inhibited anchorage-independent growth in soft agar and growth in an orthotopic xenograft model. These findings suggest a novel pathway of tumor-promoting activity by Ror2 within a subset of renal carcinomas, with significant implications for unraveling the tumorigenesis of RCC.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Proliferation , Kidney Neoplasms/pathology , Receptors, Cell Surface/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
2.
Article in English | MEDLINE | ID: mdl-19028988

ABSTRACT

In many contexts, self-renewal and differentiation of stem cells are influenced by signals from their environment, constituting a niche. It is postulated that stem cells compete for local growth factors in the niche, thereby maintaining a balance between the numbers of self-renewing and differentiated cells. A critical aspect of the niche model for stem cell regulation is that the availability of self-renewing factors is limited and that stem cells compete for these factors (Fig. 1). Consequently, the range and concentrations of the niche factors are of critical importance. Now that some of the few self-renewing factors have become identified, aspects of the niche models can be tested experimentally. In this chapter, we address mechanisms of signal regulation that take place at the level of signal-producing cells, constituting a niche for stem cells. We emphasize the biochemical properties and posttranslational modifications of the signals, all in the context of Wnt signaling. We propose that these modifications control the range of Wnt signaling and have critical roles in establishing niches for stem cells in various tissues.


Subject(s)
Drosophila/physiology , Stem Cells/cytology , Stem Cells/physiology , Wnt Proteins/physiology , Animals , Cell Differentiation , Cell Proliferation , Drosophila/cytology , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Embryonic Stem Cells , Humans , Models, Biological , Protein Processing, Post-Translational , Signal Transduction , Wnt Proteins/chemistry , Wnt Proteins/genetics
3.
Semin Cell Dev Biol ; 19(5): 434-43, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18824114

ABSTRACT

The Wnt pathway constitutes one of the most attractive candidates for modulating skeletal tissue regeneration based on its functions during skeletal development and homeostasis. Wnts participate in every stage of skeletogenesis, from the self-renewal and proliferation of skeletal stem cells to the specification of osteochondroprogenitor cells and the maturation of chondrocytes and osteoblasts. We propose that the function of Wnts depend upon a skeletogenic cell's state of differentiation. In this review we summarize recent data with a focus on the roles of Wnt signaling in mesenchymal stem cell fate, osteoprogenitor cell differentiation, chondrocyte maturation, bone remodeling, and bone regeneration.


Subject(s)
Bone Development/physiology , Bone and Bones/physiology , Physiological Phenomena/physiology , Regenerative Medicine , Wnt Proteins/metabolism , Animals , Bone Regeneration , Bone Remodeling , Bone and Bones/metabolism , Cell Differentiation , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/physiology , Humans , Mesenchymal Stem Cells/cytology , Models, Biological , Signal Transduction/genetics , Wnt Proteins/genetics
4.
Oncogene ; 25(57): 7461-8, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17143290

ABSTRACT

Cell to cell communication is vital throughout the development of multicellular organisms and during adult homeostasis. One way in which communication is achieved is through the secretion of signaling molecules that are received by neighboring responding cells. Wnt ligands comprise a large family of secreted, hydrophobic, glycoproteins that control a variety of developmental and adult processes in all metazoan organisms. By binding to various receptors present on receiving cells, Wnts initiate intracellular signaling cascades resulting in changes in gene transcription. Misregulation of Wnt signaling contributes to cancer and other degenerative disorders; thus, much effort has been made to understand the ways in which the pathway is controlled. Although ample research into the regulatory mechanisms that influence intracellular signaling events has proved fruitful, a great deal still remains to be elucidated regarding the mechanisms that control Wnt protein processing and secretion from cells, transport through the extracellular space, and protein reception on neighboring cells. This review attempts to consolidate the current data regarding these essential processes.


Subject(s)
Wnt Proteins/metabolism , Wnt Proteins/physiology , Animals , Cell Communication , Drosophila , Humans , Ligands , Lipids , Models, Biological , Signal Transduction , Transcription, Genetic
7.
Nature ; 411(6835): 325-30, 2001 May 17.
Article in English | MEDLINE | ID: mdl-11357137

ABSTRACT

The acquisition of neural fate by embryonic ectodermal cells is a fundamental step in the formation of the vertebrate nervous system. Neural induction seems to involve signalling by fibroblast growth factors (FGFs) and attenuation of the activity of bone morphogenetic protein (BMP). But FGFs, either alone or in combination with BMP antagonists, are not sufficient to induce neural fate in prospective epidermal ectoderm of amniote embryos. These findings suggest that additional signals are involved in the specification of neural fate. Here we show that the state of Wnt signalling is a critical determinant of neural and epidermal fates in the chick embryo. Continual Wnt signalling blocks the response of epiblast cells to FGF signals, permitting the expression and signalling of BMP to direct an epidermal fate. Conversely, a lack of exposure of epiblast cells to Wnt signals permits FGFs to induce a neural fate.


Subject(s)
Cell Differentiation , Cell Lineage , Epidermis/embryology , Neurons/cytology , Proto-Oncogene Proteins/physiology , Signal Transduction , Xenopus Proteins , Zebrafish Proteins , Animals , Biomarkers/analysis , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Carrier Proteins , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cells, Cultured , Chick Embryo , Ectoderm/cytology , Ectoderm/drug effects , Ectoderm/metabolism , Embryonic Induction/drug effects , Epidermal Cells , Epidermis/drug effects , Epidermis/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Immunohistochemistry , Models, Biological , Neurons/drug effects , Neurons/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Pyrroles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , Transcription Factors/analysis , Wnt Proteins
9.
Genes Dev ; 15(6): 658-71, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11274052

ABSTRACT

In Drosophila embryos the protein Naked cuticle (Nkd) limits the effects of the Wnt signal Wingless (Wg) during early segmentation. nkd loss of function results in segment polarity defects and embryonic death, but how nkd affects Wnt signaling is unknown. Using ectopic expression, we find that Nkd affects, in a cell-autonomous manner, a transduction step between the Wnt signaling components Dishevelled (Dsh) and Zeste-white 3 kinase (Zw3). Zw3 is essential for repressing Wg target-gene transcription in the absence of a Wg signal, and the role of Wg is to relieve this inhibition. Our double-mutant analysis shows that, in contrast to Zw3, Nkd acts when the Wg pathway is active to restrain signal transduction. Yeast two hybrid and in vitro experiments indicate that Nkd directly binds to the basic-PDZ region of Dsh. Specially timed Nkd overexpression is capable of abolishing Dsh function in a distinct signaling pathway that controls planar-cell polarity. Our results suggest that Nkd acts directly through Dsh to limit Wg activity and thus determines how efficiently Wnt signals stabilize Armadillo (Arm)/beta-catenin and activate downstream genes.


Subject(s)
Drosophila Proteins , Glycogen Synthase Kinase 3 , Insect Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Signal Transduction , Trans-Activators , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Animals , Armadillo Domain Proteins , Body Patterning , COS Cells , Crosses, Genetic , Dishevelled Proteins , Drosophila/genetics , Electrophoresis, Polyacrylamide Gel , Epistasis, Genetic , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , Insect Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Mutagenesis , Mutation , Phenotype , Phosphoproteins/genetics , Photoreceptor Cells, Invertebrate/metabolism , Precipitin Tests , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Transcription Factors , Two-Hybrid System Techniques , Wnt Proteins
10.
Mol Cell ; 6(1): 117-26, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10949033

ABSTRACT

The Frizzled (Fz) protein in Drosophila is a bifunctional receptor that acts through a GTPase pathway in planar polarity signaling and as a receptor for Wingless (Wg) using the canonical Wnt pathway. We found that the ligand-binding domain (CRD) of Fz has an approximately 10-fold lower affinity for Wg than the CRD of DFz2, a Wg receptor without polarity activity. When the Fz CRD is replaced by the high-affinity CRD of DFz2, the resulting chimeric protein gains Wg signaling activity, yet also retains polarity signaling activity. In contrast, the reciprocal exchange of the Fz CRD onto DFz2 is not sufficient to confer polarity activity to DFz2. This suggests that Fz has an intrinsic capacity for polarity signaling and that high-affinity interaction with Wg couples it to the Wnt pathway.


Subject(s)
Drosophila Proteins , Drosophila/growth & development , Drosophila/metabolism , Insect Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Animals , Drosophila/genetics , Female , Frizzled Receptors , Genes, Insect , Insect Proteins/chemistry , Insect Proteins/genetics , Ligands , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Wings, Animal/growth & development , Wnt1 Protein
11.
Immunity ; 13(1): 15-24, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10933391

ABSTRACT

Lymphocyte enhancer factor-1 (LEF-1) is a member of the LEF-1/TCF family of transcription factors, which have been implicated in Wnt signaling and tumorigenesis. LEF-1 was originally identified in pre-B and T cells, but its function in B lymphocyte development remains unknown. Here we report that LEF-1-deficient mice exhibit defects in pro-B cell proliferation and survival in vitro and in vivo. We further show that Lef1-/- pro-B cells display elevated levels of fas and c-myc transcription, providing a potential mechanism for their increased sensitivity to apoptosis. Finally, we establish a link between Wnt signaling and normal B cell development by demonstrating that Wnt proteins are mitogenic for pro-B cells and that this effect is mediated by LEF-1.


Subject(s)
B-Lymphocytes/cytology , DNA-Binding Proteins/metabolism , Leukopoiesis/physiology , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Apoptosis , B-Lymphocytes/metabolism , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cell Survival , DNA, Complementary , DNA-Binding Proteins/genetics , Gene Expression Regulation , Glycogen Synthase Kinase 3 , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukocyte Common Antigens/analysis , Lymphoid Enhancer-Binding Factor 1 , Mice , Mice, Knockout , Molecular Sequence Data , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Wnt Proteins , Wnt-5a Protein , Wnt3 Protein , bcl-X Protein , fas Receptor/genetics
13.
Development ; 126(18): 4165-73, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10457025

ABSTRACT

The vertebrate Axin protein, the product of the mouse fused gene, binds to beta-catenin to inhibit Wnt signaling. We have identified a homolog of Axin in Drosophila, Daxin. Using double-stranded RNA interference, we generated loss-of-function phenotypes that are similar to overexpression of the Drosophila Wnt gene wingless (wg). Overexpression of Daxin produces phenotypes similar to loss of wg. In addition, we show that Daxin overexpression can modify phenotypes elicited by wg and another Drosophila Wnt gene, DWnt-2. Using immunoprecipitation of endogenous Daxin protein from embryos we show that Daxin interacts with Armadillo and Zeste-white 3. The loss-of-function and overexpression phenotypes show that Daxin, like its mammalian counterpart, acts as a negative regulator of wg/Wnt signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/genetics , Glycogen Synthase Kinase 3 , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Signal Transduction , Trans-Activators , Amino Acid Sequence , Animals , Armadillo Domain Proteins , Axin Protein , Base Sequence , Drosophila melanogaster/embryology , Embryo, Nonmammalian , Expressed Sequence Tags , Eye/growth & development , Eye Abnormalities/genetics , Female , Gene Expression Regulation, Developmental , Genetic Techniques , Insect Proteins/genetics , Insect Proteins/metabolism , Molecular Sequence Data , Ovary/growth & development , Ovary/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA/genetics , Sequence Homology, Amino Acid , Transcription Factors , Wings, Animal/growth & development , Wnt1 Protein , Wnt2 Protein
14.
Development ; 126(18): 4175-86, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10457026

ABSTRACT

In cell culture assays, Frizzled and Dfrizzled2, two members of the Frizzled family of integral membrane proteins, are able to bind Wingless and transduce the Wingless signal. To address the role of these proteins in the intact organism and to explore the question of specificity of ligand-receptor interactions in vivo, we have conducted a genetic analysis of frizzled and Dfrizzled2 in the embryo. These experiments utilize a small gamma-ray-induced deficiency that uncovers Dfrizzled2. Mutants lacking maternal frizzled and zygotic frizzled and Dfrizzled2 exhibit defects in the embryonic epidermis, CNS, heart and midgut that are indistinguishable from those observed in wingless mutants. Epidermal patterning defects in the frizzled, Dfrizzled2 double-mutant embryos can be rescued by ectopic expression of either gene. In frizzled, Dfrizzled2 mutant embryos, ectopic production of Wingless does not detectably alter the epidermal patterning defect, but ectopic production of an activated form of Armadillo produces a naked cuticle phenotype indistinguishable from that produced by ectopic production of activated Armadillo in wild-type embryos. These experiments indicate that frizzled and Dfrizzled2 function downstream of wingless and upstream of armadillo, consistent with their proposed roles as Wingless receptors. The lack of an effect on epidermal patterning of ectopic Wingless in a frizzled, Dfrizzled2 double mutant argues against the existence of additional Wingless receptors in the embryo or a model in which Frizzled and Dfrizzled2 act simply to present the ligand to its bona fide receptor. These data lead to the conclusion that Frizzled and Dfrizzled2 function as redundant Wingless receptors in multiple embryonic tissues and that this role is accurately reflected in tissue culture experiments. The redundancy of Frizzled and Dfrizzled2 explains why Wingless receptors were not identified in earlier genetic screens for mutants defective in embryonic patterning.


Subject(s)
Drosophila Proteins , Drosophila/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Neurotransmitter/metabolism , Trans-Activators , Animals , Armadillo Domain Proteins , Body Patterning/genetics , DNA Transposable Elements , Digestive System/embryology , Drosophila/genetics , Embryo, Nonmammalian , Epidermis/embryology , Frizzled Receptors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homozygote , Insect Proteins/genetics , Insect Proteins/metabolism , Membrane Proteins/genetics , Mutation , Neurons , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt1 Protein
15.
Genes Dev ; 13(14): 1768-73, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10421629

ABSTRACT

The stabilization of beta-catenin is a key regulatory step during cell fate changes and transformations to tumor cells. Several interacting proteins, including Axin, APC, and the protein kinase GSK-3beta are implicated in regulating beta-catenin phosphorylation and its subsequent degradation. Wnt signaling stabilizes beta-catenin, but it was not clear whether and how Wnt signaling regulates the beta-catenin complex. Here we show that Axin is dephosphorylated in response to Wnt signaling. The dephosphorylated Axin binds beta-catenin less efficiently than the phosphorylated form. Thus, Wnt signaling lowers Axin's affinity for beta-catenin, thereby disengaging beta-catenin from the degradation machinery.


Subject(s)
Cytoskeletal Proteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Trans-Activators , Zebrafish Proteins , Axin Protein , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Glycogen Synthase Kinase 3 , Phosphorylation , Protein Binding , Signal Transduction , Wnt Proteins , beta Catenin
16.
Nature ; 398(6726): 431-6, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10201374

ABSTRACT

The Wnt proteins constitute a large family of extracellular signalling molecules that are found throughout the animal kingdom and are important for a wide variety of normal and pathological developmental processes. Here we describe Wnt-inhibitory factor-1 (WIF-1), a secreted protein that binds to Wnt proteins and inhibits their activities. WIF-1 is present in fish, amphibia and mammals, and is expressed during Xenopus and zebrafish development in a complex pattern that includes paraxial presomitic mesoderm, notochord, branchial arches and neural crest derivatives. We use Xenopus embryos to show that WIF-1 overexpression affects somitogenesis (the generation of trunk mesoderm segments), in agreement with its normal expression in paraxial mesoderm. In vitro, WIF-1 binds to Drosophila Wingless and Xenopus Wnt8 produced by Drosophila S2 cells. Together with earlier results obtained with the secreted Frizzled-related proteins, our results indicate that Wnt proteins interact with structurally diverse extracellular inhibitors, presumably to fine-tune the spatial and temporal patterns of Wnt activity.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cricetinae , Extracellular Matrix Proteins , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Wnt Proteins , Wnt1 Protein , Xenopus , Xenopus Proteins , Zebrafish
18.
Dev Biol ; 207(1): 133-49, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10049570

ABSTRACT

Characterization of the molecular pathways controlling differentiation and proliferation in mammalian hair follicles is central to our understanding of the regulation of normal hair growth, the basis of hereditary hair loss diseases, and the origin of follicle-based tumors. We demonstrate that the proto-oncogene Wnt3, which encodes a secreted paracrine signaling molecule, is expressed in developing and mature hair follicles and that its overexpression in transgenic mouse skin causes a short-hair phenotype due to altered differentiation of hair shaft precursor cells, and cyclical balding resulting from hair shaft structural defects and associated with an abnormal profile of protein expression in the hair shaft. A putative effector molecule for WNT3 signaling, the cytoplasmic protein Dishevelled 2 (DVL2), is normally present at high levels in a subset of cells in the outer root sheath and in precursor cells of the hair shaft cortex and cuticle which lie immediately adjacent to Wnt3-expressing cells. Overexpression of Dvl2 in the outer root sheath mimics the short-hair phenotype produced by overexpression of Wnt3, supporting the hypothesis that Wnt3 and Dvl2 have the potential to act in the same pathway in the regulation of hair growth. These experiments demonstrate a previously unrecognized role for WNT signaling in the control of hair growth and structure, as well as presenting the first example of a mammalian phenotype resulting from overexpression of a Dvl gene and providing an accessible in vivo system for analysis of mammalian WNT signaling pathways.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hair/growth & development , Proteins/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Adaptor Proteins, Signal Transducing , Animals , Cell Differentiation/genetics , Dishevelled Proteins , Electrophoresis, Gel, Two-Dimensional , Epidermis/embryology , Fluorescent Antibody Technique , Hair/cytology , Hair/ultrastructure , Hair Follicle/cytology , Hair Follicle/growth & development , Histocytochemistry , In Situ Hybridization , Mice , Mice, Transgenic , Microscopy, Electron , Phenotype , Phosphoproteins , RNA, Messenger/metabolism , Transgenes/genetics , Wnt Proteins , Wnt3 Protein
20.
Development ; 125(8): 1469-76, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9502727

ABSTRACT

Patterning of the Drosophila embryo depends on the accurate expression of wingless (wg), which encodes a secreted signal required for segmentation and many other processes. Early expression of wg is regulated by the nuclear proteins of the gap and pair-rule gene classes but, after gastrulation, wg transcription is also dependent on cell-cell communication. Signaling to the Wg-producing cells is mediated by the secreted protein, Hedgehog (Hh), and by Cubitus interruptus (Ci), a transcriptional effector of the Hh signal transduction pathway. The transmembrane protein Patched (Ptc) acts as a negative regulator of wg expression; ptc- embryos have ectopic wg expression. According to the current models, Ptc is a receptor for Hh. The default activity of Ptc is to inhibit Ci function; when Ptc binds Hh, this inhibition is released and Ci can control wg transcription. We have investigated cis-acting sequences that regulate wg during the time that wg expression depends on Hh signaling. We show that approximately 4.5 kb immediately upstream of the wg transcription unit can direct expression of the reporter gene lacZ in domains similar to the normal wg pattern in the embryonic ectoderm. Expression of this reporter construct expands in ptc mutants and responds to hh activity. Within this 4.5 kb, a 150 bp element, highly conserved between D. melanogaster and Drosophila virilis, is required to spatially restrict wg transcription. Activity of this element depends on ptc, but it contains no consensus Ci-binding sites. The discovery of an element that is likely to bind a transcriptional repressor was unexpected, since the prevailing model suggests that wg expression is principally controlled by Hh signaling acting through the Ci activator. We show that wg regulatory DNA can drive lacZ in a proper wg-like pattern without any conserved Ci-binding sites and suggest that Ci can not be the sole endpoint of the Hh pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila melanogaster/embryology , Drosophila/embryology , Embryo, Nonmammalian/physiology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Binding Sites , Conserved Sequence , Gene Expression Regulation, Developmental , Hedgehog Proteins , Insect Proteins/metabolism , Molecular Sequence Data , Sequence Alignment , Sequence Deletion , Sequence Homology, Nucleic Acid , Signal Transduction , Transcription Factors , Transcription, Genetic , Wnt1 Protein , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...