Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
2.
J Assist Reprod Genet ; 39(8): 1693-1712, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35870095

ABSTRACT

Since 2007, the Oncofertility Consortium Annual Conference has brought together a diverse network of individuals from a wide range of backgrounds and professional levels to disseminate emerging basic and clinical research findings in fertility preservation. This network also developed enduring educational materials to accelerate the pace and quality of field-wide scientific communication. Between 2007 and 2019, the Oncofertility Consortium Annual Conference was held as an in-person event in Chicago, IL. The conference attracted approximately 250 attendees each year representing 20 countries around the world. In 2020, however, the COVID-19 pandemic disrupted this paradigm and precluded an in-person meeting. Nevertheless, there remained an undeniable demand for the oncofertility community to convene. To maintain the momentum of the field, the Oncofertility Consortium hosted a day-long virtual meeting on March 5, 2021, with the theme of "Oncofertility Around the Globe" to highlight the diversity of clinical care and translational research that is ongoing around the world in this discipline. This virtual meeting was hosted using the vFairs ® conference platform and allowed over 700 people to participate, many of whom were first-time conference attendees. The agenda featured concurrent sessions from presenters in six continents which provided attendees a complete overview of the field and furthered our mission to create a global community of oncofertility practice. This paper provides a synopsis of talks delivered at this event and highlights the new advances and frontiers in the fields of oncofertility and fertility preservation around the globe from clinical practice and patient-centered efforts to translational research.


Subject(s)
COVID-19 , Fertility Preservation , Neoplasms , COVID-19/epidemiology , Humans , Pandemics
3.
Oncogene ; 25(57): 7482-91, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17143292

ABSTRACT

At the heart of the canonical Wnt signaling pathway is the beta-catenin destruction complex, which functions in the absence of Wnt signaling to keep the cytosolic and nuclear levels of beta-catenin very low by promoting the phosphorylation and ubiquitination of beta-catenin. Structural studies, combined with other experimental approaches, have begun to provide important insights into the mechanism of the destruction complex. We suggest a working model for the destruction complex based on the existing structural and experimental data, and focus on the questions that this model and other studies have raised about the function of the complex in both the normal and Wnt-inhibited states.


Subject(s)
beta Catenin/chemistry , beta Catenin/physiology , Animals , Cell Nucleus/metabolism , Colonic Neoplasms/metabolism , Cytosol/metabolism , Humans , Models, Biological , Phosphorylation , Protein Conformation , Stem Cells/metabolism , Ubiquitin/chemistry
4.
Nat Struct Biol ; 8(12): 1048-52, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713475

ABSTRACT

Accumulation of the Wnt pathway effector beta-catenin is a hallmark of a number of cancers, including colon cancer. As beta-catenin accumulates in the cell, it forms a complex with Tcf family transcription factors and activates the transcription of several critical genes involved in cell proliferation. Because Tcf4 is the predominant Tcf factor present in colon cancer cells, drugs that specifically disrupt the beta-catenin-Tcf4 complex could be useful in treating colon cancers. Earlier structural and biochemical studies demonstrated that the central region of the beta-catenin binding domain of Tcf is essential for anchoring Tcf to beta-catenin via two conserved lysines in beta-catenin (called the charged 'buttons'). Here we report the crystal structure of a beta-catenin-Tcf4 complex at 2.0 A resolution. Our structural and mutagenesis studies show that Tcf4 docks specifically to beta-catenin using several distinct conformations in its essential central region. These conformations allow different glutamate residues in the central region of Tcf4 to form a salt bridge with the same critical charged button, Lys 312 of beta-catenin. We propose that this interaction may be the first event in beta-catenin-Tcf4 recognition.


Subject(s)
Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , HMGB Proteins , Protein Interaction Mapping , Trans-Activators , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Glutamic Acid/metabolism , Humans , Lysine/metabolism , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Precipitin Tests , Protein Binding , Protein Conformation , Repetitive Sequences, Amino Acid/genetics , Static Electricity , Substrate Specificity , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transcription Factor 7-Like 2 Protein , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , beta Catenin
5.
Development ; 128(12): 2407-20, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493559

ABSTRACT

Ventralizing transcriptional repressors in the Vox/Vent family have been proposed to be important regulators of dorsoventral patterning in the early embryo. While the zebrafish genes vox (vega1) and vent (vega2) both have ventralizing activity in overexpression assays, loss-of-function studies are needed to determine whether these genes have distinct or redundant functions in dorsoventral patterning and to provide critical tests of the proposed regulatory interactions among vox, vent and other genes that act to establish the dorsoventral axis. We show that vox and vent are redundant repressors of dorsal fates in zebrafish. Mutants that lack vox function have little or no dorsoventral patterning defect, and inactivation of either vox or vent by injection of antisense morpholino oligonucleotides has little or no effect on the embryo. In contrast, embryos that lack both vox and vent function have a dorsalized phenotype. Expression of dorsal mesodermal genes, including chordin, goosecoid and bozozok, is strongly expanded in embryos that lack vox and vent function, indicating that the redundant action of vox and vent is required to restrict dorsal genes to their appropriate territories. Our genetic analysis indicates that the dorsalizing transcription factor Bozozok promotes dorsal fates indirectly, by antagonizing the expression of vox and vent. In turn, vox and vent repress chordin expression, restricting its function as an antagonist of ventral fates to the dorsal side of the embryo. Our results support a model in which BMP signaling induces the expression of ventral genes, while vox and vent act redundantly to prevent the expression of chordin, goosecoid and other dorsal genes in the lateral and ventral mesendoderm.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental , Glycoproteins , Homeodomain Proteins/physiology , Intercellular Signaling Peptides and Proteins , Repressor Proteins/physiology , Xenopus Proteins , Zebrafish Proteins , Animals , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mesoderm/physiology , Mutagenesis , Phenotype , Point Mutation , Proteins/genetics , Proteins/metabolism , Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/physiology
6.
Mol Cell ; 7(4): 801-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11336703

ABSTRACT

To understand how the Wnt coreceptor LRP-5 is involved in transducing the canonical Wnt signals, we identified Axin as a protein that interacts with the intracellular domain of LRP-5. LRP-5, when expressed in fibroblast cells, showed no effect on the canonical Wnt signaling pathway by itself, but acted synergistically with Wnt. In contrast, LRP-5 mutants lacking the extracellular domain functioned as constitutively active forms that bind Axin and that induce LEF-1 activation by destabilizing Axin and stabilizing beta-catenin. Addition of Wnt caused the translocation of Axin to the membrane and enhanced the interaction between Axin and LRP-5. In addition, the LRP-5 sequences involved in interactions with Axin are required for LEF-1 activation. Thus, we conclude that the binding of Axin to LRP-5 is an important part of the Wnt signal transduction pathway.


Subject(s)
Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, LDL/metabolism , Repressor Proteins , Signal Transduction/physiology , Zebrafish Proteins , 3T3 Cells , Amino Acid Sequence , Animals , Axin Protein , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression/physiology , Glycogen Synthase Kinase 3 , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Mammals , Mice , Molecular Sequence Data , Mutagenesis/physiology , Protein Binding/physiology , Protein Structure, Tertiary , Proteins/genetics , Proto-Oncogene Proteins/genetics , Receptors, LDL/chemistry , Receptors, LDL/genetics , Wnt Proteins
7.
Dev Biol ; 229(2): 287-306, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11203696

ABSTRACT

Nodal and Nodal-related factors play fundamental roles in a number of developmental processes, including mesoderm and endoderm formation, patterning of the anterior neural plate, and determination of bilateral asymmetry in vertebrates. pitx2, a paired-like homeobox gene, has been proposed to act downstream of Nodal in the gene cascade providing left-right cues to the developing organs. Here, we report that pitx2 is required early in the Nodal signaling pathway for specification of the endodermal and mesodermal germ layers. We found that pitx2 is expressed very early during Xenopus and zebrafish development and in many regions where Nodal signaling is required, including the presumptive mesoderm and endoderm at the blastula and gastrula stages and the prechordal mesoderm at later stages. In Xenopus embryos, overexpression of pitx2 caused ectopic expression of goosecoid and sox-17 and interfered with mesoderm formation. Overexpression of pitx2 in Xenopus animal cap explants partially mimics the effects of Nodal overexpression, suggesting that pitx2 is a mediator of Nodal signaling during specification of the endoderm and prechordal plate, but not during mesoderm induction. We further demonstrate that pitx2 is induced by Nodal signaling in Xenopus animal caps and that the early expression of zebrafish pitx2 is absent when the Nodal signaling pathway is inactive. Inhibition of pitx2 function using a chimeric EnR-pitx2 blocked specification of the mesoderm and endoderm and caused severe embryonic defects resembling those seen when Nodal signaling is inhibited. Following inhibition of pitx2 function, the fate of ventral vegetal blastomeres was shifted from an endodermal to a more mesodermal fate, an effect that was reversed by wild-type pitx2. Finally, we show that inhibition of pitx2 function interferes with the response of cells to Nodal signaling. Our results provide direct evidence that pitx2 function is required for normal specification of the endodermal and mesodermal germ layers.


Subject(s)
Embryo, Nonmammalian/physiology , Endoderm/physiology , Gastrula/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Nuclear Proteins , Signal Transduction/physiology , Transcription Factors/physiology , Transforming Growth Factor beta/physiology , Amino Acid Sequence , Animals , Cloning, Molecular , Histones/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Molecular Sequence Data , Nodal Protein , Open Reading Frames , Paired Box Transcription Factors , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Transforming Growth Factor beta/genetics , Vertebrates , Xenopus/embryology , Zebrafish/embryology , Zygote/physiology , Homeobox Protein PITX2
8.
Mech Dev ; 100(1): 93-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11118891

ABSTRACT

T-box transcription factors are important determinants of embryonic cell fate and behaviour. Two T-box genes are expressed in the developing telencephalon of several vertebrate species, including amphibia, birds and mammals. Here we report the cloning of zebrafish T-brain-1 (tbr1) and eomesodermin (eom). As a prelude to genetic studies of neuro-ectodermal fate determination we studied their expression pattern during embryogenesis and early larval development. Eom is expressed in the presumptive telencephalon from around the 4-5 somite stage in bilaterally symmetric groups of cells; the number of positive cells increases dramatically with time and encompasses the entire dorsal telencephalon by the 22 somite stage. Tbr1 is expressed from the 18 somite stage in a subset of eom-expressing cells. By 24 hpf eom and tbr1 are expressed in largely overlapping domains in the dorsal telencephalon, tbr1 is expressed in postmitotic cells whereas eomes is also expressed in proliferative ventricular zone cells. Both genes are also found in a small domain of the diencephalon bordering the telencephalon. A detailed analysis of the expression of tbr1 and eom in the brain of 4 day old larvae shows that the two T-box genes are differentially expressed in various cell populations of the developing brain.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Prosencephalon/embryology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Zebrafish Proteins , Zebrafish/embryology , Amino Acid Sequence , Animals , DNA, Complementary/metabolism , Gene Library , In Situ Hybridization , Models, Biological , Molecular Sequence Data , Olfactory Bulb/embryology , Phylogeny , Sequence Homology, Amino Acid , Time Factors
9.
Development ; 127(21): 4681-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11023870

ABSTRACT

Formation of the three germ layers requires a series of inductive events during early embryogenesis. Studies in zebrafish indicate that the source of these inductive signals may be the extra-embryonic yolk syncytial layer (YSL). The characterization of genes encoding the nodal-related factor, Squint, and homeodomain protein, Bozozok, both of which are expressed in the YSL, suggested that the YSL has a role in mesendoderm induction. However, these genes, and a second nodal-related factor, cyclops, are also expressed in the overlying marginal blastomeres, raising the possibility that the marginal blastomeres can induce mesendodermal genes independently of the YSL. We have developed a novel technique to study signaling from the YSL in which we specifically eliminate RNAs in the YSL, thus addressing the in vivo requirement of RNA-derived signals from this region in mesendoderm induction. We show that injection of RNase into the yolk cell after the 1K cell stage (3 hours) effectively eliminates YSL transcripts without affecting ubiquitously expressed genes in the blastoderm. We also present data that indicate the stability of existing proteins in the YSL is unaffected by RNase injection. Using this technique, we show that RNA in the YSL is required for the formation of ventrolateral mesendoderm and induction of the nodal-related genes in the ventrolateral marginal blastomeres, revealing the presence of an unidentified inducing signal released from the YSL. We also demonstrate that the dorsal mesoderm can be induced independently of signals from the YSL and present evidence that this is due to the stabilization of (&bgr;)-catenin in the dorsal marginal blastomeres. Our results demonstrate that germ layer formation and patterning in zebrafish uses a combination of YSL-dependent and -independent inductive events.


Subject(s)
Body Patterning , Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Mesoderm/physiology , Zebrafish Proteins , Zebrafish/embryology , Animals , Egg Yolk/cytology , Egg Yolk/physiology , Embryo, Nonmammalian/cytology , Giant Cells/cytology , Giant Cells/physiology , Homeodomain Proteins/genetics , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , Mesoderm/cytology , Nodal Protein , Nodal Signaling Ligands , Ribonucleases/metabolism , Transforming Growth Factor beta/genetics
11.
Dev Biol ; 224(2): 275-85, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10926766

ABSTRACT

Fish and frog embryos are patterned along the dorsal-ventral axis during the gastrula stage by opposing gradients of Bmps and Bmp inhibitory proteins. Three transcriptional repressors with partially overlapping expression domains have been proposed to be important mediators of Bmp function in Xenopus. We find that two related factors are expressed in the early zebrafish embryo. Although these factors are considerably divergent from the related Xenopus genes, they are expressed in domains similar to those of their Xenopus relatives throughout embryogenesis. Both of the zebrafish genes, which we have named vox and vent, are potent ventralizing factors in both zebrafish and Xenopus embryos. Using mutants in the Bmp pathway, we find that there are Bmp-dependent and Bmp-independent domains of vox expression, whereas vent is mostly dependent upon Bmp signaling. We show that ectopic vox or vent negatively regulates expression of the early dorsal gene bozozok (boz) and that ectopic boz eliminates vox and vent expression. Moreover, the normal exclusion of vox and vent from the organizer region is lost in boz mutant embryos. Our results show that boz and vox/vent are mutually antagonistic and indicate that the early establishment of the size of the organizer domain is dependent on an interplay between these early expressed transcriptional repressors.


Subject(s)
Body Patterning/genetics , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Molecular Sequence Data , Repressor Proteins/chemistry , Sequence Homology, Amino Acid , Zebrafish Proteins/chemistry
12.
Curr Opin Genet Dev ; 10(4): 350-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10889062

ABSTRACT

Many of the key molecular events underlying the induction and patterning of the vertebrate mesoderm and endoderm have recently been elucidated. T-box transcription factors and TGF-beta and Wnt signaling pathways play crucial roles in the initial induction of the mesendoderm and the subdivision of the posterior mesoderm into rostral and caudal domains.


Subject(s)
Body Patterning/physiology , Embryonic Induction/physiology , Endoderm/physiology , Mesoderm/physiology , Animals , Vertebrates/embryology , Xenopus/embryology , Zebrafish/embryology
13.
Development ; 127(10): 2143-54, 2000 May.
Article in English | MEDLINE | ID: mdl-10769238

ABSTRACT

Structurally unrelated neural inducers in vertebrate and invertebrate embryos have been proposed to function by binding to BMP4 or Dpp, respectively, and preventing these homologous signals from activating their receptor(s). In this study, we investigate the functions of various forms of the Drosophila Sog protein using the discriminating assay of Drosophila wing development. We find that misexpression of Drosophila Sog, or its vertebrate counterpart Chordin, generates a very limited vein-loss phenotype. This sog misexpression phenotype is very similar to that of viable mutants of glass-bottom boat (gbb), which encodes a BMP family member. Consistent with Sog selectively interfering with Gbb signaling, Sog can block the effect of misexpressing Gbb, but not Dpp in the wing. In contrast to the limited BMP inhibitory activity of Sog, we have identified carboxy-truncated forms of Sog, referred to as Supersog, which when misexpressed cause a broad range of dpp(-) mutant phenotypes. In line with its phenotypic effects, Supersog can block the effects of both misexpressing Dpp and Gbb in the wing. Vertebrate Noggin, on the other hand, acts as a general inhibitor of Dpp signaling, which can interfere with the effect of overexpressing Dpp, but not Gbb. We present evidence that Sog processing occurs in vivo and is biologically relevant. Overexpression of intact Sog in embryos and adult wing primordia leads to the developmentally regulated processing of Sog. This in vivo processing of Sog can be duplicated in vitro by treating Sog with a combination of the metalloprotease Tolloid (Tld) plus Twisted Gastrulation (Tsg), another extracellular factor involved in Dpp signaling. In accord with this result, coexpression of intact Sog and Tsg in developing wings generates a phenotype very similar to that of Supersog. Finally, we provide evidence that tsg functions in the embryo to generate a Supersog-like activity, since Supersog can partially rescue tsg(-) mutants. Consistent with this finding, sog(- )and tsg(-) mutants exhibit similar dorsal patterning defects during early gastrulation. These results indicate that differential processing of Sog generates a novel BMP inhibitory activity during development and, more generally, that BMP antagonists play distinct roles in regulating the quality as well as the magnitude of BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Drosophila Proteins , Glycoproteins , Insect Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carrier Proteins , Cell Line , Drosophila/growth & development , Insect Proteins/genetics , Metalloendopeptidases/metabolism , Mutagenesis , Phenotype , Proteins/genetics , Signal Transduction , Tolloid-Like Metalloproteinases , Wings, Animal/growth & development , Xenopus/embryology
14.
J Neurosci ; 20(7): 2567-74, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10729337

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) activity is negatively regulated by several signal transduction cascades that protect neurons against apoptosis, including the phosphatidylinositol-3 kinase (PI-3 kinase) pathway. This suggests the interesting possibility that activation of GSK3beta may contribute to neuronal apoptosis. Consequently, we evaluated the role of GSK3beta in apoptosis in cultured cortical neurons induced by trophic factor withdrawal or by PI-3 kinase inhibition. Neurons were subjected to several apoptotic paradigms, including serum deprivation, serum deprivation combined with exposure to NMDA receptor antagonists, or treatment with PI-3 kinase inhibitors. These treatments all led to stimulation of GSK3beta activity in cortical neurons, which preceded the induction of apoptosis. Expression of an inhibitory GSK3beta binding protein or a dominant interfering form of GSK3beta reduced neuronal apoptosis, suggesting that GSK3beta contributes to trophic factor withdrawal-induced apoptosis. Furthermore, overexpression of GSK3beta in neurons increased apoptosis, indicating that activation of this enzyme is sufficient to trigger programmed cell death. Although destabilization of beta-catenin is an important physiological effect of GSK3beta activation, expression of a mutant beta-catenin that is not destabilized by GSK3beta did not protect against apoptosis. We conclude that inhibition of GSK3beta is one of the mechanisms by which PI-3 kinase activation protects neurons from programmed cell death.


Subject(s)
Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Microtubule-Associated Proteins/physiology , Neurons/physiology , Trans-Activators , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Chromones/pharmacology , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , beta Catenin
15.
J Cell Biol ; 148(4): 691-702, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10684251

ABSTRACT

Glycogen synthase kinase 3 (GSK-3) is a constitutively active kinase that negatively regulates its substrates, one of which is beta-catenin, a downstream effector of the Wnt signaling pathway that is required for dorsal-ventral axis specification in the Xenopus embryo. GSK-3 activity is regulated through the opposing activities of multiple proteins. Axin, GSK-3, and beta-catenin form a complex that promotes the GSK-3-mediated phosphorylation and subsequent degradation of beta-catenin. Adenomatous polyposis coli (APC) joins the complex and downregulates beta-catenin in mammalian cells, but its role in Xenopus is less clear. In contrast, GBP, which is required for axis formation in Xenopus, binds and inhibits GSK-3. We show here that GSK-3 binding protein (GBP) inhibits GSK-3, in part, by preventing Axin from binding GSK-3. Similarly, we present evidence that a dominant-negative GSK-3 mutant, which causes the same effects as GBP, keeps endogenous GSK-3 from binding to Axin. We show that GBP also functions by preventing the GSK-3-mediated phosphorylation of a protein substrate without eliminating its catalytic activity. Finally, we show that the previously demonstrated axis-inducing property of overexpressed APC is attributable to its ability to stabilize cytoplasmic beta-catenin levels, demonstrating that APC is impinging upon the canonical Wnt pathway in this model system. These results contribute to our growing understanding of how GSK-3 regulation in the early embryo leads to regional differences in beta-catenin levels and establishment of the dorsal axis.


Subject(s)
Body Patterning , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Proteins/metabolism , Repressor Proteins , Trans-Activators , Xenopus Proteins , Zebrafish Proteins , Adenomatous Polyposis Coli Protein , Animals , Axin Protein , Calcium-Calmodulin-Dependent Protein Kinases/administration & dosage , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Cyclic AMP Response Element-Binding Protein/chemistry , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Enzyme Activation , Genes, Dominant/genetics , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Intracellular Signaling Peptides and Proteins , Microinjections , Models, Biological , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Proteins/administration & dosage , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins/physiology , Rats , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Signal Transduction , Wnt Proteins , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism , beta Catenin
16.
Dev Biol ; 218(2): 235-47, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10656766

ABSTRACT

T-box transcription factors are critical regulators of early embryonic development. We have characterized a novel zebrafish T-box transcription factor, hrT (H15-related T box) that is a close relative of Drosophila H15 and a recently identified human gene. We show that Drosophila H15 and zebrafish hrT are both expressed early during heart formation, in strong support of previous work postulating that vertebrate and arthropod hearts are homologous structures with conserved regulatory mechanisms. The timing and regulation of zebrafish hrT expression in anterior lateral plate mesoderm suggest a very early role for hrT in the differentiation of the cardiac precursors. hrT is coexpressed with gata4 and nkx2.5 not only in anterior lateral plate mesoderm but also in noncardiac mesoderm adjacent to the tail bud, suggesting that a conserved regulatory pathway links expression of these three genes in cardiac and noncardiac tissues. Finally, we analyzed hrT expression in pandora mutant embryos, since these have defects in many of the tissues that express hrT, including the heart. hrT expression is much reduced in the early heart fields of pandora mutants, whereas it is ectopically expressed subsequently. Using hrT expression as a marker, we describe a midline patterning defect in pandora affecting the anterior hindbrain and associated midline mesendodermal derivatives. We discuss the possibility that the cardiac ventricular defect previously described in pandora and the midline defects described here are related.


Subject(s)
Drosophila/embryology , Heart/embryology , Transcription Factors/physiology , Zebrafish/embryology , Amino Acid Sequence , Animals , Gene Expression Regulation, Developmental , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Transcription Factors/chemistry
17.
Cell ; 103(6): 885-96, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11136974

ABSTRACT

The Wnt signaling pathway plays critical roles in embryonic development and tumorigenesis. Stimulation of the Wnt pathway results in the accumulation of a nuclear beta-catenin/Tcf complex, activating Wnt target genes. A crystal structure of beta-catenin bound to the beta-catenin binding domain of Tcf3 (Tcf3-CBD) has been determined. The Tcf3-CBD forms an elongated structure with three binding modules that runs antiparallel to beta-catenin along the positively charged groove formed by the armadillo repeats. Structure-based mutagenesis defines three sites in beta-catenin that are critical for binding the Tcf3-CBD and are differentially involved in binding APC, cadherin, and Axin. The structural and mutagenesis data reveal a potential target for molecular drug design studies.


Subject(s)
Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , HMGB Proteins , Protein Conformation , Repressor Proteins , Trans-Activators , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Axin Protein , Cadherins/metabolism , Crystallography, X-Ray , Cytoskeletal Proteins/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Precipitin Tests , Protein Binding , Proteins/metabolism , Sequence Alignment , Signal Transduction , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transcription Factors/chemistry , Xenopus , Xenopus Proteins , beta Catenin
18.
Curr Opin Genet Dev ; 9(5): 553-8, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508686

ABSTRACT

Xenopus has played a key role in defining the general mechanisms that underlie early vertebrate development. Recent studies reveal how the transcriptional regulation of signaling and transcription factors is used to pattern the early dorsal-ventral axis. With the development of new methods for producing transgenic frogs, Xenopus will become a very attractive system for studying transcriptional regulation at all stages of embryogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Transcription, Genetic , Animals , Embryonic Development , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Xenopus/genetics , Xenopus Proteins
19.
EMBO J ; 18(15): 4233-40, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10428961

ABSTRACT

Wnt proteins transduce their signals through dishevelled (Dvl) proteins to inhibit glycogen synthase kinase 3beta (GSK), leading to the accumulation of cytosolic beta-catenin and activation of TCF/LEF-1 transcription factors. To understand the mechanism by which Dvl acts through GSK to regulate LEF-1, we investigated the roles of Axin and Frat1 in Wnt-mediated activation of LEF-1 in mammalian cells. We found that Dvl interacts with Axin and with Frat1, both of which interact with GSK. Similarly, the Frat1 homolog GBP binds Xenopus Dishevelled in an interaction that requires GSK. We also found that Dvl, Axin and GSK can form a ternary complex bridged by Axin, and that Frat1 can be recruited into this complex probably by Dvl. The observation that the Dvl-binding domain of either Frat1 or Axin was able to inhibit Wnt-1-induced LEF-1 activation suggests that the interactions between Dvl and Axin and between Dvl and Frat may be important for this signaling pathway. Furthermore, Wnt-1 appeared to promote the disintegration of the Frat1-Dvl-GSK-Axin complex, resulting in the dissociation of GSK from Axin. Thus, formation of the quaternary complex may be an important step in Wnt signaling, by which Dvl recruits Frat1, leading to Frat1-mediated dissociation of GSK from Axin.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Phosphoproteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins , Transcription Factors/metabolism , Zebrafish Proteins , Adaptor Proteins, Signal Transducing , Animals , Axin Protein , Dishevelled Proteins , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Lymphoid Enhancer-Binding Factor 1 , Protein Binding , Protein Conformation , Signal Transduction , Wnt Proteins , Wnt1 Protein , Xenopus , Xenopus Proteins
20.
Dev Biol ; 211(2): 293-305, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10395789

ABSTRACT

Patterning in the vertebrate embryo is controlled by an interplay between signals from the dorsal organizer and the ventrally expressed BMPs. Here we examine the function of Vox, a homeodomain-containing gene that is activated by the ventralizing signal BMP-4. Inhibition of BMP signaling using a dominant negative BMP receptor (DeltaBMPR) leads to the ectopic activation of dorsal genes in the ventral marginal zone, and this activation is prevented by co-injection of Vox. chordin is the most strongly activated of those genes that are up-regulated by DeltaBMPR and is the gene most strongly inhibited by Vox expression. We demonstrate that Vox acts as a transcriptional repressor, showing that the activity of native Vox is mimicked by a Vox-repressor fusion (VoxEnR) and that a Vox-activator fusion (VoxG4A) acts as an antimorph, causing the formation of a partial secondary axis when expressed on the ventral side of the embryo. Although Vox can ectopically activate BMP-4 expression in whole embryos, we see no activation of BMP-4 by VoxG4A, demonstrating that this activation is indirect. Using a hormone-inducible version of VoxG4A, we find that a critical time window for Vox function is during the late blastula period. Using this construct, we demonstrate that only a subset of dorsal genes is directly repressed by Vox, revealing that there are different modes of regulation for organizer genes. Since the major direct target for Vox repression is chordin, we propose that Vox acts in establishing a BMP-4 morphogen gradient by restricting the expression domain of chordin.


Subject(s)
Bone Morphogenetic Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/physiology , Xenopus Proteins , Xenopus/embryology , Xenopus/physiology , Animals , Bone Morphogenetic Protein 4 , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...