370 results on '"Basler, K"'
Search Results
102. A genome‐wide RNA interference screen uncovers two p24 proteins as regulators of Wingless secretion
- Author
-
Fillip Port, Konrad Basler, George Hausmann, University of Zurich, and Basler, K
- Subjects
animal structures ,1303 Biochemistry ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,1311 Genetics ,RNA interference ,1312 Molecular Biology ,Genetics ,Secretion ,Receptor ,Molecular Biology ,Gene ,Tissue homeostasis ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Endoplasmic reticulum ,fungi ,Scientific Reports ,Wnt signaling pathway ,10124 Institute of Molecular Life Sciences ,Cell biology ,chemistry ,570 Life sciences ,biology ,Glycoprotein ,030217 neurology & neurosurgery - Abstract
Wnt proteins are secreted, lipid-modified glycoproteins that control animal development and adult tissue homeostasis. Secretion of Wnt proteins is at least partly regulated by a dedicated machinery. Here, we report a genome-wide RNA interference screen for genes involved in the secretion of Wingless (Wg), a Drosophila Wnt. We identify three new genes required for Wg secretion. Of these, Emp24 and Eclair are required for proper export of Wg from the endoplasmic reticulum (ER). We propose that Emp24 and Eca act as specific cargo receptors for Wg to concentrate it in forming vesicles at sites of ER export.
- Published
- 2011
- Full Text
- View/download PDF
103. The role of Parafibromin/Hyrax as a nuclear Gli/Ci-interacting protein in Hedgehog target gene control
- Author
-
Christian Mosimann, Konrad Basler, George Hausmann, University of Zurich, and Basler, K
- Subjects
Transcriptional Activation ,Embryology ,Carboxy-Lyases ,Parafibromin ,Genes, Insect ,Zinc Finger Protein GLI1 ,Cell Line ,1309 Developmental Biology ,Genes, Reporter ,GLI1 ,GLI2 ,Animals ,Drosophila Proteins ,Wings, Animal ,Hedgehog Proteins ,Hedgehog ,Transcription factor ,Conserved Sequence ,Tissue homeostasis ,Cell Nucleus ,Oncogene Proteins ,Genetics ,Gene knockdown ,biology ,Gene Expression Regulation, Developmental ,2710 Embryology ,10124 Institute of Molecular Life Sciences ,Hedgehog signaling pathway ,Cell biology ,DNA-Binding Proteins ,Drosophila melanogaster ,Trans-Activators ,biology.protein ,570 Life sciences ,RNA Interference ,Protein Binding ,Signal Transduction ,Transcription Factors ,Developmental Biology - Abstract
The Hedgehog (Hh) pathway, an evolutionarily conserved key regulator of embryonic patterning and tissue homeostasis, controls its target genes by managing the processing and activities of the Gli/Ci transcription factors. Little is known about the nuclear co-factors the Gli/Ci proteins recruit, and how they mechanistically control Hh target genes. Here, we provide evidence for the involvement of Parafibromin/Hyx as a positive component in Hh signaling. We found that hyx RNAi impaired Hh pathway activity in Drosophila cell culture. Consistent with an evolutionarily conserved function in Hh signaling, RNAi-mediated knockdown of Parafibromin in mammalian cell culture experiments diminished the transcriptional activity of Gli1 and Gli2. In vivo , in Drosophila , genetic impairment of hyx decreased the expression of the high-threshold Hh target gene knot / collier . Conversely, hyx overexpression ameliorated the inhibitory activity of Ptc and Ci 75 misexpression during Drosophila wing development. We subsequently found that Parafibromin can form a complex with all three Glis, and provide evidence that Parafibromin/Hyx directly binds Region 1, the Su(fu) interaction domain, in the N-terminus of all Glis and Ci. Taken together, our results suggest a target gene-selective involvement of the PAF1 complex in Hh signaling via the Parafibromin/Hyx-mediated recruitment to Gli/Ci.
- Published
- 2009
- Full Text
- View/download PDF
104. Wingless secretion promotes and requires retromer-dependent cycling of Wntless
- Author
-
George Hausmann, Fillip Port, Konrad Basler, Carla Bänziger, Patrick Herr, Edy Furger, Marco Kuster, University of Zurich, and Basler, K
- Subjects
Retromer ,Vesicular Transport Proteins ,Golgi Apparatus ,Wnt1 Protein ,Biology ,Endocytosis ,1307 Cell Biology ,symbols.namesake ,Proto-Oncogene Proteins ,Animals ,Drosophila Proteins ,Secretion ,chemistry.chemical_classification ,Intracellular Signaling Peptides and Proteins ,Wnt signaling pathway ,Cell Biology ,Golgi apparatus ,10124 Institute of Molecular Life Sciences ,Transmembrane protein ,Cell biology ,Wnt Proteins ,Retromer complex ,Protein Transport ,chemistry ,symbols ,570 Life sciences ,biology ,Drosophila ,Glycoprotein - Abstract
Wnt ligands are lipid-modified, secreted glycoproteins that control multiple steps during embryogenesis and adult-tissue homeostasis. Little is known about the mechanisms underlying Wnt secretion. Recently, Wntless (Wls/Evi/Srt) was identified as a conserved multi-pass transmembrane protein whose function seems to be dedicated to promoting the release of Wnts. Here, we describe Wls accumulation in the Golgi apparatus of Wnt/Wingless (Wg)-producing cells in Drosophila, and show that this localization is essential for Wg secretion. Moreover, Wls localization and levels critically depend on retromer, a conserved protein complex that mediates endosome-to-Golgi protein trafficking in yeast. In the absence of the retromer components Dvps35 or Dvps26, but in presence of Wg, Wls is degraded and Wg secretion impaired. Our results indicate that Wg, clathrin-mediated endocytosis and retromer sustain a Wls traffic loop from the Golgi to the plasma membrane and back to the Golgi, thereby enabling Wls to direct Wnt secretion.
- Published
- 2008
- Full Text
- View/download PDF
105. Helping Wingless take flight: how WNT proteins are secreted
- Author
-
George Hausmann, Konrad Basler, Carla Bänziger, University of Zurich, and Basler, K
- Subjects
Lipoproteins ,Golgi Apparatus ,Endosomes ,Biology ,Models, Biological ,1307 Cell Biology ,1312 Molecular Biology ,Animals ,Humans ,Secretion ,Molecular Biology ,Wnt signaling pathway ,Membrane Proteins ,LRP6 ,LRP5 ,Cell Biology ,Wnt secretion ,10124 Institute of Molecular Life Sciences ,Transmembrane protein ,Cell biology ,Wnt Proteins ,Retromer complex ,Protein Transport ,Biochemistry ,570 Life sciences ,biology ,Signal Transduction ,Lipoprotein - Abstract
How functional WNT proteins are made and how their secretion is regulated is becoming a focal point for the WNT-signalling field. Recently, lipoprotein particles, WNT lipid modifications, the conserved transmembrane protein Wntless (WLS; also known as EVI and SRT) and the retromer complex have been implicated in WNT secretion. Our aim is to synthesize ideas from these new findings for the mechanisms that underlie WNT secretion.
- Published
- 2007
- Full Text
- View/download PDF
106. An ancient defense system eliminates unfit cells from developing tissues during cell competition
- Author
-
S N Meyer, Marc Amoyel, Laura A. Johnston, C de la Cova, Konrad Basler, Cora Bergantiños, Claus Schertel, University of Zurich, and Basler, K
- Subjects
Cell signaling ,Transcription, Genetic ,medicine.medical_treatment ,Cell ,Population ,Apoptosis ,Cell Communication ,Biology ,medicine ,Animals ,Drosophila Proteins ,education ,Transcription factor ,Gene ,Genetics ,education.field_of_study ,1000 Multidisciplinary ,Multidisciplinary ,Innate immune system ,Neuropeptides ,Toll-Like Receptors ,NF-kappa B ,biology.organism_classification ,Immunity, Innate ,10124 Institute of Molecular Life Sciences ,Cell biology ,Drosophila melanogaster ,Cytokine ,medicine.anatomical_structure ,Mutation ,570 Life sciences ,biology ,Transcription Factors - Abstract
Developing tissues that contain mutant or compromised cells present risks to animal health. Accordingly, the appearance of a population of suboptimal cells in a tissue elicits cellular interactions that prevent their contribution to the adult. Here we report that this quality control process, cell competition, uses specific components of the evolutionarily ancient and conserved innate immune system to eliminate Drosophila cells perceived as unfit. We find that Toll-related receptors (TRRs) and the cytokine Spätzle (Spz) lead to NFκB-dependent apoptosis. Diverse “loser” cells require different TRRs and NFκB factors and activate distinct pro-death genes, implying that the particular response is stipulated by the competitive context. Our findings demonstrate a functional repurposing of components of TRRs and NFκB signaling modules in the surveillance of cell fitness during development.
- Published
- 2014
- Full Text
- View/download PDF
107. Opposing Transcriptional Outputs of Hedgehog Signaling and Engrailed Control Compartmental Cell Sorting at the Drosophila A/P Boundary
- Author
-
Christian Dahmann, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Transcription, Genetic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Animals, Genetically Modified ,Transformation, Genetic ,1300 General Biochemistry, Genetics and Molecular Biology ,Cell Movement ,Cell Adhesion ,Animals ,Drosophila Proteins ,Hedgehog Proteins ,Hedgehog ,Genetics ,Homeodomain Proteins ,Activator (genetics) ,Cell adhesion molecule ,Biochemistry, Genetics and Molecular Biology(all) ,Ci protein ,Cell sorting ,engrailed ,Hedgehog signaling pathway ,10124 Institute of Molecular Life Sciences ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Imaginal disc ,Trans-Activators ,570 Life sciences ,biology ,Insect Proteins ,Drosophila ,Signal Transduction ,Transcription Factors - Abstract
The wing imaginal disc is subdivided into two nonintermingling sets of cells, the anterior (A) and posterior (P) compartments. Anterior cells require reception of the Hedgehog (Hh) signal to segregate from P cells. We provide evidence that Hh signaling controls A/P cell segregation not by directly modifying structural components but by a Cubitus interruptus (Ci)-mediated transcriptional response. A shift in the balance between repressor and activator forms of Ci toward the activator form is necessary and sufficient to define “A-type” cell sorting behavior. Moreover, we show that Engrailed (En), in the absence of Ci, is sufficient to specify “P-type” sorting. We propose that the opposing transcriptional activities of Ci and En control cell segregation at the A/P boundary by regulating a single cell adhesion molecule.
- Published
- 2000
- Full Text
- View/download PDF
108. Comment on 'Dynamics of dpp signaling and proliferation control'
- Author
-
Simon Restrepo, Schu-Fee Yang, Konrad Basler, Gerald Schwank, University of Zurich, and Basler, K
- Subjects
0303 health sciences ,1000 Multidisciplinary ,animal structures ,Multidisciplinary ,Wing ,biology ,Dynamics (mechanics) ,biology.organism_classification ,10124 Institute of Molecular Life Sciences ,Cell biology ,03 medical and health sciences ,Drosophila melanogaster ,0302 clinical medicine ,SX00 SystemsX.ch ,SX15 WingX ,Animals ,Drosophila Proteins ,Intercellular Signaling Peptides and Proteins ,Wings, Animal ,570 Life sciences ,Drosophila (subgenus) ,030217 neurology & neurosurgery ,Cell Proliferation ,Signal Transduction ,030304 developmental biology - Abstract
Wartlick et al . (Research Articles, 4 March 2011, p. 1154) reported that growth rates in the Drosophila wing disc correlate with increasing Dpp signaling levels, suggesting that the rate of Dpp increase determines the cell-cycle length. Contradicting their model, we found that cells in which the increase of Dpp signaling levels was genetically abrogated grew at rates comparable to those of wild-type cells.
- Published
- 2012
- Full Text
- View/download PDF
109. Compartment boundarles and the control of Drosophila limb pattern by bedgebog protein
- Author
-
Basler, K. and Struhl, G.
- Published
- 1994
- Full Text
- View/download PDF
110. Probing transcription-specific outputs of β-catenin in vivo
- Author
-
Tomas Valenta, Max Gay, Sarah Steiner, Kalina Draganova, Martina Zemke, Raymond Hoffmans, Paolo Cinelli, Michel Aguet, Lukas Sommer, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Beta-catenin ,10017 Institute of Anatomy ,610 Medicine & health ,Mice, Inbred Strains ,1309 Developmental Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,1311 Genetics ,Genetics ,medicine ,Animals ,10239 Institute of Laboratory Animal Science ,Cell adhesion ,Wnt Signaling Pathway ,beta Catenin ,030304 developmental biology ,0303 health sciences ,biology ,Gastrulation ,Wnt signaling pathway ,Neural tube ,LRP6 ,Gene Expression Regulation, Developmental ,LRP5 ,Epithelial Cells ,Adherens Junctions ,Cell biology ,Wnt Proteins ,medicine.anatomical_structure ,Spinal Cord ,Catenin ,Mesoderm formation ,Mutation ,Perspective ,biology.protein ,570 Life sciences ,030217 neurology & neurosurgery ,Research Paper ,Developmental Biology ,Signal Transduction - Abstract
β-Catenin, apart from playing a cell-adhesive role, is a key nuclear effector of Wnt signaling. Based on activity assays in Drosophila, we generated mouse strains where the endogenous β-catenin protein is replaced by mutant forms, which retain the cell adhesion function but lack either or both of the N- and the C-terminal transcriptional outputs. The C-terminal activity is essential for mesoderm formation and proper gastrulation, whereas N-terminal outputs are required later during embryonic development. By combining the double-mutant β-catenin with a conditional null allele and a Wnt1-Cre driver, we probed the role of Wnt/β-catenin signaling in dorsal neural tube development. While loss of β-catenin protein in the neural tube results in severe cell adhesion defects, the morphology of cells and tissues expressing the double-mutant form is normal. Surprisingly, Wnt/β-catenin signaling activity only moderately regulates cell proliferation, but is crucial for maintaining neural progenitor identity and for neuronal differentiation in the dorsal spinal cord. Our model animals thus allow dissecting signaling and structural functions of β-catenin in vivo and provide the first genetic tool to generate cells and tissues that entirely and exclusively lack canonical Wnt pathway activity.
- Published
- 2011
111. Antagonistic growth regulation by Dpp and Fat drives uniform cell proliferation
- Author
-
Konrad Basler, Gerardo Tauriello, Gerald Schwank, Elizabeth Kranz, Petros Koumoutsakos, Ryohei Yagi, University of Zurich, and Basler, K
- Subjects
animal structures ,Growth regulation ,medicine.medical_treatment ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,1309 Developmental Biology ,1307 Cell Biology ,03 medical and health sciences ,0302 clinical medicine ,Mediator ,SX00 SystemsX.ch ,SX15 WingX ,1300 General Biochemistry, Genetics and Molecular Biology ,1312 Molecular Biology ,medicine ,Animals ,Drosophila Proteins ,Wings, Animal ,Primordium ,10. No inequality ,Molecular Biology ,030304 developmental biology ,Body Patterning ,Cell Proliferation ,0303 health sciences ,Cell growth ,Growth factor ,Cell Polarity ,Gene Expression Regulation, Developmental ,Cell Biology ,Subcellular localization ,10124 Institute of Molecular Life Sciences ,Cell biology ,Drosophila melanogaster ,Biochemistry ,570 Life sciences ,biology ,Signal transduction ,Cell Adhesion Molecules ,030217 neurology & neurosurgery ,Developmental Biology ,Morphogen ,Signal Transduction - Abstract
We use the Dpp morphogen gradient in the Drosophila wing disc as a model to address the fundamental question of how a gradient of a growth factor can produce uniform growth. We first show that proper expression and subcellular localization of components in the Fat tumor-suppressor pathway, which have been argued to depend on Dpp activity differences, are not reliant on the Dpp gradient. We next analyzed cell proliferation in discs with uniformly high Dpp or uniformly low Fat signaling activity and found that these pathways regulate growth in a complementary manner. While the Dpp mediator Brinker inhibits growth in the primordium primarily in the lateral regions, Fat represses growth mostly in the medial region. Together, our results indicate that the activities of both signaling pathways are regulated in a parallel rather than sequential manner and that uniform proliferation is achieved by their complementary action on growth.
- Published
- 2011
- Full Text
- View/download PDF
112. Coop functions as a corepressor of Pangolin and antagonizes Wingless signaling
- Author
-
Haiyun Song, Johannes Bischof, Sandra Goetze, Erich Brunner, Konrad Basler, Chloe Spichiger-Haeusermann, Marco Kuster, University of Zurich, and Basler, K
- Subjects
animal structures ,Repressor ,Wnt1 Protein ,1309 Developmental Biology ,03 medical and health sciences ,Research Communication ,1311 Genetics ,Transcription (biology) ,biology.animal ,Genetics ,Animals ,Drosophila Proteins ,Transcription factor ,Psychological repression ,030304 developmental biology ,Regulation of gene expression ,Armadillo Domain Proteins ,0303 health sciences ,biology ,integumentary system ,030302 biochemistry & molecular biology ,Pangolin ,biology.organism_classification ,10124 Institute of Molecular Life Sciences ,Repressor Proteins ,Drosophila melanogaster ,Gene Expression Regulation ,Armadillo ,embryonic structures ,570 Life sciences ,Corepressor ,Co-Repressor Proteins ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
Wingless (Wg) signaling regulates expression of its target genes via Pangolin and Armadillo, and their interacting cofactors. In the absence of Wg, Pangolin mediates transcriptional repression. In the presence of Wg, Pangolin, Armadillo, and a cohort of coactivators mediate transcriptional activation. Here we uncover Coop (corepressor of Pan) as a Pangolin-interacting protein. Coop and Pangolin form a complex on DNA containing a Pangolin/TCF-binding motif. Overexpression of Coop specifically represses Wg target genes, while loss of Coop function causes derepression. Finally, we show that Coop antagonizes the binding of Armadillo to Pangolin, providing a mechanism for Coop-mediated repression of Wg target gene transcription.
- Published
- 2010
113. Exploring the effects of mechanical feedback on epithelial topology
- Author
-
Konrad Basler, Tinri Aegerter-Wilmsen, Christof M. Aegerter, Alix J. Christen, Alister C. Smith, Ernst Hafen, University of Zurich, and Basler, K
- Subjects
Cell division ,530 Physics ,Cell ,Population ,Mitosis ,10192 Physics Institute ,Apical cell ,Biology ,Topology ,1309 Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,SX00 SystemsX.ch ,SX15 WingX ,1312 Molecular Biology ,medicine ,Animals ,Wings, Animal ,Computer Simulation ,education ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Cell Size ,0303 health sciences ,education.field_of_study ,Wing ,Cell growth ,Epithelial Cells ,Models, Theoretical ,10124 Institute of Molecular Life Sciences ,medicine.anatomical_structure ,Polygon ,570 Life sciences ,biology ,Drosophila ,Stress, Mechanical ,U7 Systems Biology / Functional Genomics ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Apical cell surfaces in metazoan epithelia, such as the wing disc of Drosophila, resemble polygons with different numbers of neighboring cells. The distribution of these polygon numbers has been shown to be conserved. Revealing the mechanisms that lead to this topology might yield insights into how the structural integrity of epithelial tissues is maintained. It has previously been proposed that cell division alone, or cell division in combination with cell rearrangements, is sufficient to explain the observed epithelial topology. Here, we extend this work by including an analysis of the clustering and the polygon distribution of mitotic cells. In addition, we study possible effects of cellular growth regulation by mechanical forces, as such regulation has been proposed to be involved in wing disc size regulation. We formulated several theoretical scenarios that differ with respect to whether cell rearrangements are allowed and whether cellular growth rates are dependent on mechanical stress. We then compared these scenarios with experimental data on the polygon distribution of the entire cell population, that of mitotic cells, as well as with data on mitotic clustering. Surprisingly, we observed considerably less clustering in our experiments than has been reported previously. Only scenarios that include mechanical-stress-dependent growth rates are in agreement with the experimental data. Interestingly, simulations of these scenarios showed a large decrease in rearrangements and elimination of cells. Thus, a possible growth regulation by mechanical force could have a function in releasing the mechanical stress that evolves when all cells have similar growth rates.
- Published
- 2010
114. Refined LexA transactivators and their use in combination with the Drosophila Gal4 system
- Author
-
Konrad Basler, Franz Mayer, Ryohei Yagi, University of Zurich, and Basler, K
- Subjects
animal structures ,Repressor ,Computational biology ,Biology ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,SX00 SystemsX.ch ,Genes, Reporter ,SX15 WingX ,Animals ,Drosophila Proteins ,Wings, Animal ,Enhancer trap ,Transcription factor ,030304 developmental biology ,Genetics ,1000 Multidisciplinary ,0303 health sciences ,Multidisciplinary ,fungi ,Biological Sciences ,biology.organism_classification ,10124 Institute of Molecular Life Sciences ,Repressor Proteins ,Drosophila melanogaster ,Enhancer Elements, Genetic ,Trans-Activators ,570 Life sciences ,biology ,Repressor lexA ,030217 neurology & neurosurgery ,Drosophila Protein ,Signal Transduction ,Transcription Factors - Abstract
The use of binary transcriptional systems offers many advantages for experimentally manipulating gene activity, as exemplified by the success of the Gal4/UAS system in Drosophila . To expand the number of applications, a second independent transactivator (TA) is desirable. Here, we present the optimization of an additional system based on LexA and show how it can be applied. We developed a series of LexA TAs, selectively suppressible via Gal80, that exhibit high transcriptional activity and low detrimental effects when expressed in vivo. In combination with Gal4, an appropriately selected LexA TA permits to program cells with a distinct balance and independent outputs of the two TAs. We demonstrate how the two systems can be combined for manipulating communicating cell populations, converting transient tissue-specific expression patterns into heritable, constitutive activities, and defining cell territories by intersecting TA expression domains. Finally, we describe a versatile enhancer trap system that allows swapping TA and generating mosaics composed of Gal4 and LexA TA-expressing cells. The optimized LexA system facilitates precise analyses of complex biological phenomena and signaling pathways in Drosophila .
- Published
- 2010
- Full Text
- View/download PDF
115. Transcription in the absence of histone H3.3
- Author
-
Martina Hödl, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Histone H3 Lysine 4 ,Transcription, Genetic ,Blotting, Western ,DEVBIO ,1100 General Agricultural and Biological Sciences ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Histone H3 ,1300 General Biochemistry, Genetics and Molecular Biology ,Histone methylation ,Histone H2A ,Animals ,Histone code ,Cloning, Molecular ,DNA Primers ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Reproduction ,Pioneer factor ,DNA ,Immunohistochemistry ,Molecular biology ,Chromatin ,10124 Institute of Molecular Life Sciences ,Histone methyltransferase ,Mutation ,H3K4me3 ,570 Life sciences ,biology ,Drosophila ,General Agricultural and Biological Sciences ,Signal Transduction - Abstract
SummaryDi- and trimethylation of histone H3 lysine 4 (H3K4me2 and H3K4me3) are hallmarks of chromatin at active genes [1]. The major fraction of K4-methylated histone H3 is the variant H3 (termed H3.3 in Drosophila) [2], which replaces canonical H3 (H3.2) in transcribed genes [3, 4]. Here, we genetically address the in vivo significance of such K4 methylation by replacing wild-type H3.3 with a mutant form (H3.3K4A) that cannot be methylated. We monitored the transcription that occurs in response to multiple well-described signaling pathways. Surprisingly, the transcriptional outputs of these pathways remain intact in H3.3K4A mutant cells. Even the complete absence of both H3.3 genes does not noticeably affect viability or function of cells: double mutant animals are viable but sterile. Fertility can be rescued by K4-containing versions of H3.3, but not with mutant H3.3 (H3.3K4A) or with canonical H3.2. Together, these data suggest that in Drosophila, presence of H3.3K4me in the chromatin of active genes is dispensable for successful transcription in most cells and only plays an important role in reproductive tissues.
- Published
- 2009
- Full Text
- View/download PDF
116. Growth regulation by Dpp: an essential role for Brinker and a non-essential role for graded signaling levels
- Author
-
Gerald Schwank, Konrad Basler, Simon Restrepo, University of Zurich, and Basler, K
- Subjects
animal structures ,Growth regulation ,Cell ,Genes, Insect ,Organ development ,Biology ,Animals, Genetically Modified ,1309 Developmental Biology ,medicine ,1312 Molecular Biology ,Animals ,Drosophila Proteins ,Wings, Animal ,Molecular Biology ,Psychological repression ,Body Patterning ,Cell Proliferation ,Decapentaplegic ,Cell growth ,Anatomy ,10124 Institute of Molecular Life Sciences ,Cell biology ,Repressor Proteins ,Imaginal disc ,medicine.anatomical_structure ,Homogeneous ,Mutation ,570 Life sciences ,biology ,Drosophila ,Developmental Biology ,Signal Transduction - Abstract
Morphogens can control organ development by regulating patterning as well as growth. Here we use the model system of the Drosophila wing imaginal disc to address how the patterning signal Decapentaplegic (Dpp)regulates cell proliferation. Contrary to previous models, which implicated the slope of the Dpp gradient as an essential driver of cell proliferation, we find that the juxtaposition of cells with differential pathway activity is not required for proliferation. Additionally, our results demonstrate that, as is the case for patterning, Dpp controls wing growth entirely via repression of the target gene brinker (brk). The Dpp-Brk system converts an inherently uneven growth program, with excessive cell proliferation in lateral regions and low proliferation in medial regions, into a spatially homogeneous profile of cell divisions throughout the disc.
- Published
- 2008
117. Wntless, a conserved membrane protein dedicated to the secretion of Wnt proteins from signaling cells
- Author
-
Peder Zipperlen, George Hausmann, Konrad Basler, Davide Soldini, Corina Schütt, Carla Bänziger, University of Zurich, and Basler, K
- Subjects
Male ,Blastomeres ,DNA, Complementary ,Molecular Sequence Data ,Cell Communication ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Evolution, Molecular ,Wnt3 Protein ,1300 General Biochemistry, Genetics and Molecular Biology ,Wnt protein secretion ,Wnt3A Protein ,Animals ,Drosophila Proteins ,Humans ,Secretion ,Amino Acid Sequence ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Conserved Sequence ,Body Patterning ,Base Sequence ,Biochemistry, Genetics and Molecular Biology(all) ,Wnt signaling pathway ,Intracellular Signaling Peptides and Proteins ,LRP6 ,Cell Polarity ,Membrane Proteins ,LRP5 ,10124 Institute of Molecular Life Sciences ,Cell biology ,Wnt Proteins ,Drosophila melanogaster ,Membrane protein ,570 Life sciences ,biology ,Female ,Signal transduction ,Signal Transduction - Abstract
SummaryCell-cell communication via Wnt signals represents a fundamental means by which animal development and homeostasis are controlled. The identification of components of the Wnt pathway is reaching saturation for the transduction process in receiving cells but is incomplete concerning the events occurring in Wnt-secreting cells. Here, we describe the discovery of a novel Wnt pathway component, Wntless (Wls/Evi), and show that it is required for Wingless-dependent patterning processes in Drosophila, for MOM-2-governed polarization of blastomeres in C. elegans, and for Wnt3a-mediated communication between cultured human cells. In each of these cases, Wls is acting in the Wnt-sending cells to promote the secretion of Wnt proteins. Since loss of Wls function has no effect on other signaling pathways yet appears to impede all the Wnt signals we analyzed, we propose that Wls represents an ancient partner for Wnts dedicated to promoting their secretion into the extracellular milieu.
- Published
- 2005
118. Dissecting nuclear Wingless signalling: recruitment of the transcriptional co-activator Pygopus by a chain of adaptor proteins
- Author
-
Konrad Basler, Reto Städeli, University of Zurich, and Basler, K
- Subjects
Embryology ,Amino Acid Motifs ,Molecular Sequence Data ,Repressor ,Wnt1 Protein ,Biology ,Cell Line ,1309 Developmental Biology ,Proto-Oncogene Proteins ,Animals ,Drosophila Proteins ,Amino Acid Sequence ,Transcription factor ,Alleles ,Adaptor Proteins, Signal Transducing ,Armadillo Domain Proteins ,Activator (genetics) ,Wnt signaling pathway ,Intracellular Signaling Peptides and Proteins ,Signal transducing adaptor protein ,Promoter ,2710 Embryology ,biology.organism_classification ,Molecular biology ,10124 Institute of Molecular Life Sciences ,Repressor Proteins ,Drosophila melanogaster ,570 Life sciences ,biology ,Signal transduction ,Pygopus ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
Members of the Wingless (Wg)/Wnt family of secreted glycoproteins control cell fate during embryonic development and adult homeostasis. Wnt signals regulate the expression of target genes by activating a conserved signal transduction pathway. Upon receptor activation, the signal is transmitted intracellularly by stabilization of Armadillo (Arm)/beta-catenin. Arm/beta-catenin translocates to the nucleus, interacts with DNA-binding factors of the Pangolin (Pan)/TCF/LEF class and activates transcription of target genes in cooperation with the recently identified proteins Legless/BCL9 (Lgs) and Pygopus (Pygo). Here, we analyse the mode of action of Pan, Arm, Lgs, and Pygo in Drosophila cultured cells. We provide evidence that together these four proteins form a 'chain of adaptors' linking the NH2-terminal homology domain (NHD) of Pygo to the DNA-binding domain of Pan. We show that the NHD has potent transcriptional activation capacity, which differs from that of acidic activator domains and depends on a conserved NPF tripeptide. A single point mutation within this NPF motif abolishes the transcriptional activity of the Pygo NHD in vitro and strongly reduces Wg signalling in vivo. Together, our results suggest that the transcriptional output of Wg pathway activity largely relies on a 'chain of adaptors' design to direct the Pygo NHD to Wg target promoters in an Arm-dependent manner.
- Published
- 2005
119. Identification and in vivo role of the Armadillo-Legless interaction
- Author
-
Raymond Hoffmans, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Sensitivity and Specificity ,1309 Developmental Biology ,Transduction (genetics) ,biology.animal ,Neoplasms ,Two-Hybrid System Techniques ,1312 Molecular Biology ,Animals ,Drosophila Proteins ,Humans ,Transgenes ,Cloning, Molecular ,Molecular Biology ,Gene ,beta Catenin ,Genetics ,Armadillo Domain Proteins ,Binding Sites ,biology ,Wnt signaling pathway ,Signal transducing adaptor protein ,biology.organism_classification ,Phenotype ,10124 Institute of Molecular Life Sciences ,Cytoskeletal Proteins ,Armadillo repeats ,Armadillo ,Mutation ,Disease Progression ,Trans-Activators ,570 Life sciences ,Drosophila ,Pygopus ,Developmental Biology ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
The Wnt signalling system controls many fundamental processes during animal development and its deregulation has been causally linked to colorectal cancer. Transduction of Wnt signals entails the association of beta-catenin with nuclear TCF DNA-binding factors and the subsequent activation of target genes. Using genetic assays in Drosophila, we have recently identified a presumptive adaptor protein, Legless (Lgs), that binds to beta-catenin and mediates signalling activity by recruiting the transcriptional activator Pygopus (Pygo). Here, we characterize the beta-catenin/Lgs interaction and show: (1) that it is critically dependent on two acidic amino acid residues in the first Armadillo repeat of beta-catenin; (2) that it is spatially and functionally separable from the binding sites for TCF factors, APC and E-cadherin; (3) that it is required in endogenous as well as constitutively active forms of beta-catenin for Wingless signalling output in Drosophila; and (4) that in its absence animals develop with the same phenotypic consequences as animals lacking Lgs altogether. Based on these findings, and because Lgs and Pygo have human homologues that can substitute for their Drosophila counterparts, we infer that the beta-catenin/Lgs binding site may thus serve as an attractive drug target for therapeutic intervention in beta-catenin-dependent cancer progression.
- Published
- 2004
120. dMyc transforms cells into super-competitors
- Author
-
Konrad Basler, Eduardo Moreno, University of Zurich, and Basler, K
- Subjects
Cell Survival ,Organogenesis ,Cell ,Apoptosis ,Cell Communication ,Myc proteins ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,1300 General Biochemistry, Genetics and Molecular Biology ,Morphogenesis ,medicine ,Animals ,Drosophila Proteins ,Wings, Animal ,Survival signaling ,Regulation of gene expression ,Genetics ,Biochemistry, Genetics and Molecular Biology(all) ,Gene Expression Regulation, Developmental ,Cell Differentiation ,10124 Institute of Molecular Life Sciences ,Clone Cells ,Cell biology ,DNA-Binding Proteins ,Imaginal disc ,Drosophila melanogaster ,medicine.anatomical_structure ,Competitive behavior ,Bone Morphogenetic Proteins ,570 Life sciences ,biology ,Carcinogenesis ,Cell Division ,Signal Transduction ,Transcription Factors - Abstract
Overexpression of myc protooncogenes has been implicated in the genesis of many human tumors. Myc proteins seem to regulate diverse biological processes, but their role in tumorigenesis remains enigmatic. Here we use Drosophila imaginal discs to mimic situations in which cells with unequal levels of Myc protein are apposed and show that this invariably elicits a win/lose situation reminiscent of cell competition; cells with lower levels of dMyc are eliminated by apoptosis whereas cells with higher levels of dMyc overproliferate. We find that this competitive behavior correlates with, and can be corrected by, the activation of the BMP/Dpp survival signaling pathway. Hence the heritable increase in dMyc levels causes cells to behave as “super-competitors” and reveals a novel mode of clonal expansion that causes, but also relies on, the killing of surrounding cells.
- Published
- 2004
- Full Text
- View/download PDF
121. A genetic screen for hedgehog targets involved in the maintenance of the Drosophila anteroposterior compartment boundary
- Author
-
Mátyás Végh, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Patched ,Genetics ,Membrane Proteins ,Receptors, Cell Surface ,Biology ,Cell sorting ,Smoothened Receptor ,engrailed ,10124 Institute of Molecular Life Sciences ,Receptors, G-Protein-Coupled ,Imaginal disc ,1311 Genetics ,Mutation ,Animals ,Drosophila Proteins ,Wings, Animal ,570 Life sciences ,biology ,Drosophila ,Hedgehog Proteins ,Hedgehog ,Gene ,Drosophila Protein ,Genetic screen ,Research Article - Abstract
The development of multicellular organisms requires the establishment of cell populations with different adhesion properties. In Drosophila, a cell-segregation mechanism underlies the maintenance of the anterior (A) and posterior (P) compartments of the wing imaginal disc. Although engrailed (en) activity contributes to the specification of the differential cell affinity between A and P cells, recent evidence suggests that cell sorting depends largely on the transduction of the Hh signal in A cells. The activator form of Cubitus interruptus (Ci), a transcription factor mediating Hh signaling, defines anterior specificity, indicating that Hh-dependent cell sorting requires Hh target gene expression. However, the identity of the gene(s) contributing to distinct A and P cell affinities is unknown. Here, we report a genetic screen based on the FRT/FLP system to search for genes involved in the correct establishment of the anteroposterior compartment boundary. By using double FRT chromosomes in combination with a wing-specific FLP source we screened 250,000 mutagenized chromosomes. Several complementation groups affecting wing patterning have been isolated, including new alleles of most known Hh-signaling components. Among these, we identified a class of patched (ptc) alleles exhibiting a novel phenotype. These results demonstrate the value of our setup in the identification of genes involved in distinct wing-patterning processes.
- Published
- 2003
- Full Text
- View/download PDF
122. Wingful, an extracellular feedback inhibitor of wingless
- Author
-
Konrad Basler, Offer Gerlitz, University of Zurich, and Basler, K
- Subjects
animal structures ,Molecular Sequence Data ,Wnt1 Protein ,Biology ,medicine.disease_cause ,1309 Developmental Biology ,Research Communication ,1311 Genetics ,Proto-Oncogene Proteins ,Genetics ,medicine ,Extracellular ,Animals ,Drosophila Proteins ,Amino Acid Sequence ,Feedback, Physiological ,Mutation ,Sequence Homology, Amino Acid ,Esterases ,Gene Expression Regulation, Developmental ,Dally ,Molecular biology ,10124 Institute of Molecular Life Sciences ,Notum ,Cell biology ,Extracellular Matrix ,Naked cuticle ,Larva ,570 Life sciences ,biology ,Insect Proteins ,Drosophila ,Signal transduction ,Drosophila Protein ,Developmental Biology ,Morphogen ,Signal Transduction - Abstract
Secreted peptide signals control many fundamental processes during animal development. Proper responses to these signals require cognate inducible feedback antagonists. Here we report the identification of a novel Drosophila Wingless (Wg) target gene, wingful(wf), and show that it encodes a potent extracellular feedback inhibitor of Wg. In contrast to the cytoplasmic protein Naked cuticle (Nkd), the only known Wg feedback antagonist, Wf functions during larval stages, when Nkd function is dispensable. We propose that Wf may provide feedback control for the long-range morphogen activities of Wg.
- Published
- 2002
- Full Text
- View/download PDF
123. The Wingless target gene Dfz3 encodes a new member of the Drosophila Frizzled family
- Author
-
Konrad Basler, Manolo Calleja, Rajeev Sivasankaran, Ginés Morata, University of Zurich, and Basler, K
- Subjects
Frizzled ,Embryology ,animal structures ,Molecular Sequence Data ,Genes, Insect ,Receptors, Cell Surface ,Wnt1 Protein ,Biology ,Receptors, G-Protein-Coupled ,1309 Developmental Biology ,Proto-Oncogene Proteins ,Animals ,Drosophila Proteins ,Amino Acid Sequence ,Receptor ,Gene ,Peptide sequence ,Genetics ,LRP6 ,2710 Embryology ,Frizzled Receptors ,10124 Institute of Molecular Life Sciences ,Imaginal disc ,Signalling ,Insect Proteins ,570 Life sciences ,biology ,Drosophila ,Drosophila Protein ,Developmental Biology - Abstract
Here we report the identification of Dfz3, a novel member of the Frizzled family of seven-pass transmembrane receptors. Like Dfz2, Dfz3 is a target gene of Wingless (Wg) signalling, but in contrast to Dfz2, it is activated rather than repressed by Wg signalling in imaginal discs. We show that Dfz3 is not required for viability but is necessary for optimal Wg signalling at the wing margin.
- Published
- 2000
124. Direct transcriptional control of the Dpp target omb by the DNA binding protein Brinker
- Author
-
M. Alessandra Vigano, Bruno Müller, Konrad Basler, Markus Affolter, Rajeev Sivasankaran, University of Zurich, and Basler, K
- Subjects
animal structures ,Transcription, Genetic ,Response element ,Repressor ,Genes, Insect ,Nerve Tissue Proteins ,Genetics and Molecular Biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,1300 General Biochemistry, Genetics and Molecular Biology ,2400 General Immunology and Microbiology ,Transcriptional regulation ,1312 Molecular Biology ,Animals ,Drosophila Proteins ,Wings, Animal ,Binding site ,Enhancer ,Transcription factor ,Molecular Biology ,DNA Primers ,Binding Sites ,Base Sequence ,Decapentaplegic ,General Immunology and Microbiology ,General Neuroscience ,2800 General Neuroscience ,DNA ,Articles ,Molecular biology ,10124 Institute of Molecular Life Sciences ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,Mutation ,General Biochemistry ,Insect Proteins ,570 Life sciences ,biology ,Drosophila ,T-Box Domain Proteins ,Transcription Factors ,Morphogen - Abstract
The gradient morphogen Decapentaplegic (Dpp) organizes pattern by inducing the transcription of different target genes at distinct threshold concentrations during Drosophila development. An important, albeit indirect, mode by which Dpp controls the spatial extent of its targets is via the graded downregulation of brinker, whose product in turn negatively regulates the expression of these targets. Here we report the molecular dissection of the cis-regulatory sequences of optomotor-blind (omb), a Dpp target gene in the wing. We identify a minimal 284 bp Dpp response element and demonstrate that it is subject to Brinker (Brk) repression. Using this omb wing enhancer, we show that Brk is a sequence-specific DNA binding protein. Mutations in the high-affinity Brk binding site abolish responsiveness of this omb enhancer to Brk and also compromise the input of an unknown transcriptional activator. Our results therefore identify Brk as a novel transcription factor antagonizing Dpp signalling by directly binding target genes and repressing their expression.
- Published
- 2000
- Full Text
- View/download PDF
125. Dispatched, a novel sterol-sensing domain protein dedicated to the release of cholesterol-modified hedgehog from signaling cells
- Author
-
Kirsten André Senti, Konrad Basler, Manolo Bellotto, Barry J. Dickson, Denise Nellen, Ernst Hafen, Richard Burke, University of Zurich, and Basler, K
- Subjects
Patched ,Embryo, Nonmammalian ,Transgene ,Protein domain ,Molecular Sequence Data ,Receptors, Cell Surface ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Downregulation and upregulation ,1300 General Biochemistry, Genetics and Molecular Biology ,medicine ,Animals ,Drosophila Proteins ,Hedgehog Proteins ,Amino Acid Sequence ,Transgenes ,Cloning, Molecular ,Receptor ,Hedgehog ,Genetics ,Mutation ,Biochemistry, Genetics and Molecular Biology(all) ,Membrane Proteins ,Immunohistochemistry ,10124 Institute of Molecular Life Sciences ,Cell biology ,Cholesterol ,Larva ,Insect Proteins ,570 Life sciences ,biology ,Drosophila ,Sequence Alignment ,Cytoneme ,Signal Transduction - Abstract
Members of the Hedgehog (Hh) family of secreted signaling proteins function as potent short-range organizers in animal development. Their range of action is limited by a C-terminal cholesterol tether and the upregulation of Patched (Ptc) receptor levels. Here we identify a novel segment-polarity gene in Drosophila, dispatched (disp), and demonstrate that its product is required in sending cells for normal Hh function. In the absence of Disp, cholesterol-modified but not cholesterol-free Hh is retained in these cells, indicating that Disp functions to release cholesterol-anchored Hh. Despite their opposite roles, Disp and Ptc share structural homology in the form of a sterol-sensing domain, suggesting that release and sequestration of cholesterol-modified Hh may be based on related molecular pathways.
- Published
- 1999
- Full Text
- View/download PDF
126. Distinct and regulated activities of human Gli proteins in Drosophila
- Author
-
Christian von Mering, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Transcription, Genetic ,Xenopus Proteins ,Animals, Genetically Modified ,Genes, Reporter ,Genes, Synthetic ,Morphogenesis ,Drosophila Proteins ,Wings, Animal ,Sonic hedgehog ,Promoter Regions, Genetic ,Regulation of gene expression ,Oncogene Proteins ,biology ,integumentary system ,Agricultural and Biological Sciences(all) ,Gene Expression Regulation, Developmental ,10124 Institute of Molecular Life Sciences ,DNA-Binding Proteins ,Drosophila melanogaster ,embryonic structures ,Insect Proteins ,General Agricultural and Biological Sciences ,Drosophila Protein ,DNA, Complementary ,animal structures ,Recombinant Fusion Proteins ,Kruppel-Like Transcription Factors ,Repressor ,Nerve Tissue Proteins ,1100 General Agricultural and Biological Sciences ,Zinc Finger Protein GLI1 ,General Biochemistry, Genetics and Molecular Biology ,Species Specificity ,GLI1 ,Zinc Finger Protein Gli3 ,1300 General Biochemistry, Genetics and Molecular Biology ,GLI2 ,GLI3 ,Animals ,Humans ,Hedgehog Proteins ,Hedgehog ,Biochemistry, Genetics and Molecular Biology(all) ,Genetic Complementation Test ,fungi ,Proteins ,Molecular biology ,Repressor Proteins ,biology.protein ,Trans-Activators ,570 Life sciences ,Transcription Factors - Abstract
In both vertebrates and Drosophila, limb development is organized by a posteriorly located source of the signalling protein Hedgehog (Hh) [1] [2] [3] [4]. In Drosophila, the expression of Hh target genes is controlled by two opposing activities of the transcriptional regulator Cubitus interruptus (Ci), which activates target genes in response to Hh signalling but is converted into a repressor form in the absence of Hh [5] [6] [7] [8] [9] [10]. Three homologs of Ci (Gli1, Gli2, and Gli3) have been implicated in mediating responses to Sonic hedgehog (Shh) in vertebrates [11] [12]. Much attention has been devoted to the expression pattern of GLI genes; GLI1 is induced by Shh, whereas GLI3 transcription appears to be repressed by Shh signalling [13] [14] [15]. The regulation of GLI gene expression is therefore one important mechanism by which GLI genes organize pattern. It is not well understood, however, whether Shh signalling also controls the activities of Gli proteins post-translationally and whether these activities have activating or repressing effects on target genes in vivo. Here, we have subjected the human proteins Gli1 and Gli3 to the precise and well-defined Hh signalling assay of Drosophila wing development and established that Gli1 functions as an activator and Gli3 as a repressor of Hh target genes; that the activating transcriptional activity of Gli1 and the repressing activity of Gli3 are both subject to Hh regulation in vivo; and that the combined activities of Gli1 and Gli3 can substitute for Ci in controlling Hh target gene expression during embryonic and larval development.
- Published
- 1999
- Full Text
- View/download PDF
127. Drosophila ciD encodes a hybrid Pangolin/Cubitus interruptus protein that diverts the Wingless into the Hedgehog signaling pathway
- Author
-
Konrad Basler, Liang Schweizer, University of Zurich, and Basler, K
- Subjects
Patched ,Embryology ,Embryo, Nonmammalian ,Mutant ,Molecular Sequence Data ,Receptors, Cell Surface ,Wnt1 Protein ,Biology ,1309 Developmental Biology ,Proto-Oncogene Proteins ,Morphogenesis ,Animals ,Drosophila Proteins ,Wings, Animal ,Hedgehog Proteins ,Amino Acid Sequence ,Transgenes ,Promoter Regions, Genetic ,Hedgehog ,Transcription factor ,In Situ Hybridization ,Genetics ,Base Sequence ,Drosophila embryogenesis ,Gene Expression Regulation, Developmental ,Membrane Proteins ,Ci protein ,2710 Embryology ,Fusion protein ,Hedgehog signaling pathway ,10124 Institute of Molecular Life Sciences ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Drosophila melanogaster ,Genes ,Chromosome Inversion ,570 Life sciences ,biology ,Insect Proteins ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
The Hedgehog (Hh) and Wingless (Wg) signaling pathways play important roles in animal development. The activities of the two pathways depend on each other during Drosophila embryogenesis. In the embryonic segment, Wg is required in anterior cells to sustain Hh secretion in adjacent posterior cells. Hh input in turn is necessary for anterior cells to maintain wg expression. The Hh and Wg pathways are mediated by the transcription factors Cubitus interruptus (Ci) and Pangolin/TCF (Pan), respectively. Coincidentally, pan and ci are adjacent genes on the fourth chromosome in a head-to-head orientation. Our genetic and in situ hybridization data indicate that ci D is a mutation affecting both ci and pan . Molecular analysis revealed that the ci D allele is caused by an inversion event that swapped the promoter regions and the first exons of the two genes. The ci gene in ci D is controlled by the ubiquitous pan promoter and encodes a hybrid Ci protein that carries the N-terminal region of Pan. This domain has previously been shown to bind to the β-catenin homolog Armadillo (Arm), raising the possibility that Wg input, in addition to Hh input, modulates the activity of the hybrid Ci D protein. Indeed, we found that Wg signaling induces the expression of the Hh target gene patched ( ptc ) in ci D animals. We provide evidence that this effect depends on the ability of the Ci D protein to bind Arm. Genetic and molecular data indicate that wild-type Pan and Ci D compete for binding to Arm, leading to a compromised transduction of the Wg signal in heterozygous ci D /+ animals and to a dramatic enhancement of the gain-of-function activity of Ci D in homozygous mutants. Thus, the Hh and the Wg pathways are affected by the ci D mutation, and the Ci D fusion protein integrates the activities of both.
- Published
- 1998
128. Formation of the Long Range Dpp Morphogen Gradient
- Author
-
Ryohei Yagi, Konrad Basler, Markus Affolter, Gerald Schwank, Sven Bergmann, Schu-Fee Yang, Sascha Dalessi, Aitana Morton de Lachapelle, University of Zurich, and Basler, K
- Subjects
Activin Receptors, Type II ,Mutant ,0302 clinical medicine ,SX00 SystemsX.ch ,2400 General Immunology and Microbiology ,Molecular Cell Biology ,Morphogenesis ,Drosophila Proteins ,Wings, Animal ,Biology (General) ,Receptor ,Internalization ,media_common ,Genetics ,0303 health sciences ,General Neuroscience ,2800 General Neuroscience ,10124 Institute of Molecular Life Sciences ,Cell biology ,Imaginal disc ,Drosophila melanogaster ,Transcytosis ,Organ Specificity ,Larva ,General Agricultural and Biological Sciences ,Algorithms ,Research Article ,Morphogen ,animal structures ,QH301-705.5 ,media_common.quotation_subject ,Receptors, Cell Surface ,1100 General Agricultural and Biological Sciences ,Protein Serine-Threonine Kinases ,Biology ,Endocytosis ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,SX15 WingX ,1300 General Biochemistry, Genetics and Molecular Biology ,Animals ,030304 developmental biology ,General Immunology and Microbiology ,Decapentaplegic ,Computational Biology ,Activin Receptors, Type II/genetics ,Activin Receptors, Type II/metabolism ,Drosophila Proteins/genetics ,Drosophila Proteins/metabolism ,Drosophila melanogaster/anatomy & histology ,Drosophila melanogaster/genetics ,Larva/genetics ,Larva/growth & development ,Models, Chemical ,Mutation ,Protein-Serine-Threonine Kinases/genetics ,Protein-Serine-Threonine Kinases/metabolism ,Receptors, Cell Surface/genetics ,Receptors, Cell Surface/metabolism ,Wing/growth & development ,Wing/metabolism ,570 Life sciences ,biology ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The TGF-β homolog Decapentaplegic (Dpp) acts as a secreted morphogen in the Drosophila wing disc, and spreads through the target tissue in order to form a long range concentration gradient. Despite extensive studies, the mechanism by which the Dpp gradient is formed remains controversial. Two opposing mechanisms have been proposed: receptor-mediated transcytosis (RMT) and restricted extracellular diffusion (RED). In these scenarios the receptor for Dpp plays different roles. In the RMT model it is essential for endocytosis, re-secretion, and thus transport of Dpp, whereas in the RED model it merely modulates Dpp distribution by binding it at the cell surface for internalization and subsequent degradation. Here we analyzed the effect of receptor mutant clones on the Dpp profile in quantitative mathematical models representing transport by either RMT or RED. We then, using novel genetic tools, experimentally monitored the actual Dpp gradient in wing discs containing receptor gain-of-function and loss-of-function clones. Gain-of-function clones reveal that Dpp binds in vivo strongly to the type I receptor Thick veins, but not to the type II receptor Punt. Importantly, results with the loss-of-function clones then refute the RMT model for Dpp gradient formation, while supporting the RED model in which the majority of Dpp is not bound to Thick veins. Together our results show that receptor-mediated transcytosis cannot account for Dpp gradient formation, and support restricted extracellular diffusion as the main mechanism for Dpp dispersal. The properties of this mechanism, in which only a minority of Dpp is receptor-bound, may facilitate long-range distribution., Author Summary Morphogens are signaling molecules that trigger specific responses in cells in a concentration-dependent manner. The formation of morphogen gradients is essential for the patterning of tissues and organs. Decapentaplegic (Dpp) is the Drosophila homolog of the bone morphogenic proteins in vertebrates and forms a morphogen gradient along the anterior-posterior axis of the Drosophila wing imaginal disc, a single-cell layered epithelium. Dpp determines the growth and final size of the wing disc and serves as an ideal model system to study gradient formation. Despite extensive studies the mechanism by which morphogen gradients are established remains controversial. In the case of Dpp two mechanisms have been postulated, namely extracellular diffusion and receptor-mediated transcytosis. In the first model Dpp is suggested to move by diffusion through the extracellular matrix of a tissue, whereas in the latter model Dpp is transported through the cells by receptor-mediated uptake and re-secretion. In this work we combined novel genetic tools with mathematical modeling to discriminate between the two models. Our results suggest that the Dpp gradient forms following the extracellular diffusion mechanism. Moreover, our data suggest that the majority of the extracellular Dpp is free and not bound to its receptor, a property likely to play a role for the long-range gradient formation.
- Published
- 2011
- Full Text
- View/download PDF
129. Ligand-independent activation of the sevenless receptor tyrosine kinase changes the fate of cells in the developing Drosophila eye
- Author
-
Konrad Basler, Bea Christen, Ernst Hafen, University of Zurich, and Basler, K
- Subjects
Molecular Sequence Data ,Receptor Protein-Tyrosine Kinases ,Biology ,Cell fate determination ,Eye ,General Biochemistry, Genetics and Molecular Biology ,Photoreceptor cell ,Receptor tyrosine kinase ,1300 General Biochemistry, Genetics and Molecular Biology ,Drosophilidae ,medicine ,Animals ,Drosophila Proteins ,Photoreceptor Cells ,Tyrosine ,Eye Proteins ,Neurons ,Genetics ,Membrane Glycoproteins ,Base Sequence ,Stem Cells ,Membrane Proteins ,Protein-Tyrosine Kinases ,biology.organism_classification ,10124 Institute of Molecular Life Sciences ,Cell biology ,Enhancer Elements, Genetic ,medicine.anatomical_structure ,Gene Expression Regulation ,Genes ,Mutagenesis ,Multigene Family ,biology.protein ,570 Life sciences ,biology ,Drosophila ,Neural development ,Drosophila Protein - Abstract
Cell fate in the developing eye is determined by a cascade of inductive interactions. In this process, the sevenless protein--a receptor tyrosine kinase--is required for the specification of the R7 photoreceptor cell fate. We have constructed a gain-of-function sevenless mutation (SevS11) by overexpressing a truncated sevenless protein in the cells where sevenless is normally expressed. In SevS11 mutant flies, all sevenless-expressing cells initiate neural development. This results in the formation of multiple R7-like photoreceptors per ommatidium. Therefore, sevenless activity appears to be necessary and sufficient for the determination of R7 cell fate. These results illustrate the central role receptor tyrosine kinases can play in the specification of cell fate during development.
- Published
- 1991
- Full Text
- View/download PDF
130. Hedgehog Controls Limb Development by Regulating the Activities of Distinct Transcriptional Activator and Repressor Forms of Cubitus interruptus
- Author
-
Nathalie Méthot, Konrad Basler, University of Zurich, and Basler, K
- Subjects
Transcription, Genetic ,Repressor ,Biology ,General Biochemistry, Genetics and Molecular Biology ,1300 General Biochemistry, Genetics and Molecular Biology ,Transcription (biology) ,Endopeptidases ,Gene expression ,Animals ,Drosophila Proteins ,Limb development ,Hedgehog Proteins ,Transgenes ,Gene ,Hedgehog ,Alleles ,Homeodomain Proteins ,Genetics ,Biochemistry, Genetics and Molecular Biology(all) ,Activator (genetics) ,Gene Expression Regulation, Developmental ,Extremities ,Ci protein ,10124 Institute of Molecular Life Sciences ,DNA-Binding Proteins ,Repressor Proteins ,Trans-Activators ,Insect Proteins ,570 Life sciences ,biology ,Drosophila ,Transcription Factors - Abstract
Hedgehog (Hh) proteins play diverse organizing roles in development by regulating gene expression in responding cells. The Gli homolog Cubitus interruptus (Ci) is involved in controlling the transcription of Hh target genes. A repressor form of Ci arises in the absence of Hh signaling by proteolytic cleavage of intact Ci. We show that this cleavage is essential for limb patterning and is regulated by Hh in vivo. We provide evidence for the existence of a distinct activator form of Ci, which does not arise by mere prevention of Ci proteolysis, but rather depends on a separate regulatory step subject to Hh control. These different activities of Ci regulate overlapping but distinct subsets of Hh target genes. Thus, limb development is organized by the integration of different transcriptional outputs of Hh signaling.
- Full Text
- View/download PDF
131. The repressor and activator forms of Cubitus interruptus control Hedgehog target genes through common generic Gli-binding sites
- Author
-
Konrad Basler, Barbara Müller, University of Zurich, and Basler, K
- Subjects
Patched ,Molecular Sequence Data ,Repressor ,Regulatory Sequences, Nucleic Acid ,Biology ,Response Elements ,Zinc Finger Protein GLI1 ,1309 Developmental Biology ,Animals, Genetically Modified ,1312 Molecular Biology ,Animals ,Drosophila Proteins ,Wings, Animal ,Hedgehog Proteins ,Enhancer ,Molecular Biology ,Hedgehog ,Gene ,Oncogene Proteins ,Genetics ,Binding Sites ,Activator (genetics) ,Gene Expression Regulation, Developmental ,Ci protein ,10124 Institute of Molecular Life Sciences ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Imaginal disc ,Enhancer Elements, Genetic ,Trans-Activators ,570 Life sciences ,biology ,Insect Proteins ,Drosophila ,Transcription Factors ,Developmental Biology - Abstract
The Drosophila Gli homolog Cubitus interruptus (Ci) controls the transcription of Hedgehog (Hh) target genes. A repressor form of Ci arises in the absence of Hh signalling by proteolytic cleavage of intact Ci, whereas an activator form of Ci is generated in response to the Hh signal. These different activities of Ci regulate overlapping but distinct subsets of Hh target genes. To investigate the mechanisms by which the two activities of Ci exert their opposite transcriptional effect, we dissect here the imaginal disc enhancer of the dpp gene, which responds to both activities of Ci. Within a minimal disc enhancer, we identify the DNA sequences that are necessary and sufficient for the control by Ci, show that the same sequences respond to the activator and repressor forms of Ci, and demonstrate that their activities can be replaced by a single synthetic Gli-binding site. We further show that the enhancer sequences of patched, a gene responding only to the activator form of Ci, effectively integrate also the repressor activity of Ci if placed into a dpp context. These results provide in vivo evidence against the employment of distinct binding sites for the different forms of Ci and suggest that target genes responding to only one form must have acquired distant cis-regulatory elements for their selective behavior.
132. Suppressor of fused opposes Hedgehog signal transduction by impeding nuclear accumulation of the activator form of Cubitus interruptus
- Author
-
Konrad Basler, Nathalie Méthot, University of Zurich, and Basler, K
- Subjects
Repressor ,Fluorescent Antibody Technique ,Kinesins ,Biology ,Protein Serine-Threonine Kinases ,Models, Biological ,law.invention ,1309 Developmental Biology ,law ,Genes, Reporter ,1312 Molecular Biology ,Animals ,Drosophila Proteins ,Hedgehog Proteins ,Phosphorylation ,Molecular Biology ,Hedgehog ,Transcription factor ,Cell Nucleus ,Kinase ,Activator (genetics) ,Gene Expression Regulation, Developmental ,Ci protein ,Molecular biology ,Cyclic AMP-Dependent Protein Kinases ,Immunohistochemistry ,10124 Institute of Molecular Life Sciences ,DNA-Binding Proteins ,Repressor Proteins ,Protein Transport ,Drosophila melanogaster ,Mutation ,570 Life sciences ,biology ,Suppressor ,Insect Proteins ,Signal transduction ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
Hedgehog controls the expression of key developmental genes through the conversion of the transcription factor Cubitus interruptus (Ci) into either an activator (Ci[act]) or a repressor (Ci[rep]) form. Proteolytic cleavage of full-length Ci is important for the generation of Ci[rep], but little is known about how Ci[act] arises in response to Hh. Here we examine Hh signal transduction components for their role in the conversion of full-length Ci into either Ci[act] or Ci[rep]. We report that Cos2, PKA and Fused are necessary for the generation of Ci[rep], whereas the inhibition of either Cos2 or PKA activity is a prerequisite for Ci[act] formation. Fused (Fu) kinase stimulates a constitutively active form of Ci in a Hh-dependent manner, suggesting that Fu enhances the activity rather than the formation of Ci[act]. Su(fu) reduces the nuclear accumulation of the constitutively active form of Ci, arguing that Su(fu) can function subsequent to Ci[act] formation. We propose that Hh induces target gene expression by a two-step mechanism in which Ci[act] is first formed and then accumulates in the nucleus via Fu-induced neutralization of Su(fu) activity.
133. Interventions to improve primary healthcare in rural settings: A scoping review.
- Author
-
Aubrey-Basler K, Bursey K, Pike A, Penney C, Furlong B, Howells M, Al-Obaid H, Rourke J, Asghari S, and Hall A
- Subjects
- Humans, Rural Health Services organization & administration, Health Services Accessibility, Primary Health Care, Rural Population
- Abstract
Background: Residents of rural areas have poorer health status, less healthy behaviours and higher mortality than urban dwellers, issues which are commonly addressed in primary care. Strengthening primary care may be an important tool to improve the health status of rural populations., Objective: Synthesize and categorize studies that examine interventions to improve rural primary care., Eligibility Criteria: Experimental or observational studies published between January 1, 1996 and December 2022 that include an historical or concurrent control comparison., Sources of Evidence: Pubmed, CINAHL, Cochrane Library, Embase., Charting Methods: We extracted and charted data by broad category (quality, access and efficiency), study design, country of origin, publication year, aim, health condition and type of intervention studied. We assigned multiple categories to a study where relevant., Results: 372 papers met our inclusion criteria, divided among quality (82%), access (20%) and efficiency (13%) categories. A majority of papers were completed in the USA (40%), Australia (15%), China (7%) or Canada (6%). 35 (9%) papers came from countries in Africa. The most common study design was an uncontrolled before-and-after comparison (32%) and only 24% of studies used randomized designs. The number of publications each year has increased markedly over the study period from 1-2/year in 1997-99 to a peak of 49 papers in 2017., Conclusions: Despite substantial inequity in health outcomes associated with rural living, very little attention is paid to rural primary care in the scientific literature. Very few studies of rural primary care use randomized designs., Competing Interests: KAB and JR are family physicians who previously practiced in rural areas. KAB, JR, and SA are rural health services researchers with an interest in the equity of health service distribution. The authors have no other disclosures to report., (Copyright: © 2024 Aubrey-Basler et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
134. The fate of secretory cells during intestinal homeostasis, regeneration, and tumor formation is regulated by Tcf4.
- Author
-
Janeckova L, Stastna M, Hrckulak D, Berkova L, Kubovciak J, Onhajzer J, Kriz V, Dostalikova S, Mullerova T, Vecerkova K, Tenglerova M, Coufal S, Kostovcikova K, Blumberg RS, Filipp D, Basler K, Valenta T, Kolar M, and Korinek V
- Abstract
The single-layer epithelium of the gastrointestinal tract is a dynamically renewing tissue that ensures nutrient absorption, secretory and barrier functions and is involved in immune responses. The basis for this homeostatic renewal is the Wnt signaling pathway. Blocking this pathway can lead to epithelial damage, while its abnormal activation can result in the development of intestinal tumors. In this study, we investigated the dynamics of intestinal epithelial cells and tumorigenesis using a conditional mouse model. Using single-cell and bulk RNA sequencing and histological analysis, we elucidated the cellular responses following the loss of specific cell types. We focused on the fate of cells in the lower parts of the intestinal crypts and the development of colon adenomas. By partially inactivating the transcription factor Tcf4, a key effector of the Wnt signaling pathway, we analyzed the regeneration of isolated hyperproliferative foci (crypts). Our results suggest that the damaged epithelium is not restored by a specific regeneration program associated with oncofetal gene production, but rather by a standard homeostatic renewal pathway. Moreover, disruption of Tcf4 in secretory progenitors resulted in a significant shift in the cell lineage from Paneth cells to goblet cells, characterized by morphological changes and loss of Paneth cell-specific genes. We also found that hyperactivation of the Wnt signaling pathway in colonic adenomas correlated with the upregulation of genes typical of Paneth cells in the intestine, followed by the emergence of secretory tumor cells producing the Wnt3 ligand. The absence of Tcf4 led to a phenotypic shift of the tumor cells towards goblet cells. Our study presents a new model of epithelial regeneration based on the genetically driven partial elimination of intestinal crypts. We highlight the critical role of Tcf4 in the control of cell lineage decisions in the intestinal epithelium and colon tumors.
- Published
- 2024
- Full Text
- View/download PDF
135. The β-catenin C terminus links Wnt and sphingosine-1-phosphate signaling pathways to promote vascular remodeling and atherosclerosis.
- Author
-
Oliveira-Paula GH, Liu S, Maira A, Ressa G, Ferreira GC, Quintar A, Jayakumar S, Almonte V, Parikh D, Valenta T, Basler K, Hla T, Riascos-Bernal DF, and Sibinga NES
- Subjects
- Humans, beta Catenin metabolism, Vascular Remodeling, Signal Transduction, Catenins metabolism, Atherosclerosis, Lysophospholipids, Sphingosine analogs & derivatives
- Abstract
Canonical Wnt and sphingosine-1-phosphate (S1P) signaling pathways are highly conserved systems that contribute to normal vertebrate development, with key consequences for immune, nervous, and cardiovascular system function; despite these functional overlaps, little is known about Wnt/β-catenin-S1P cross-talk. In the vascular system, both Wnt/β-catenin and S1P signals affect vessel maturation, stability, and barrier function, but information regarding their potential coordination is scant. We report an instance of functional interaction between the two pathways, including evidence that S1P receptor 1 (S1PR1) is a transcriptional target of β-catenin. By studying vascular smooth muscle cells and arterial injury response, we find a specific requirement for the β-catenin carboxyl terminus, which acts to induce S1PR1, and show that this interaction is essential for vascular remodeling. We also report that pharmacological inhibition of the β-catenin carboxyl terminus reduces S1PR1 expression, neointima formation, and atherosclerosis. These findings provide mechanistic understanding of how Wnt/β-catenin and S1P systems collaborate during vascular remodeling and inform strategies for therapeutic manipulation.
- Published
- 2024
- Full Text
- View/download PDF
136. Inactivation of the tumor suppressor gene Apc synergizes with H. pylori to induce DNA damage in murine gastric stem and progenitor cells.
- Author
-
He J, Nascakova Z, Leary P, Papa G, Valenta T, Basler K, and Müller A
- Subjects
- Animals, Humans, Mice, DNA Damage, Genes, Tumor Suppressor, Receptors, G-Protein-Coupled genetics, Stem Cells, Helicobacter Infections genetics, Helicobacter Infections microbiology, Helicobacter pylori genetics, Stomach Neoplasms pathology
- Abstract
Helicobacter pylori infection is a major risk factor for the development of gastric cancer. The bacteria reside in close proximity to gastric surface mucous as well as stem and progenitor cells. Here, we take advantage of wild-type and genetically engineered murine gastric organoids and organoid-derived monolayers to study the cellular targets of H. pylori -induced DNA damage and replication stress and to explore possible interactions with preexisting gastric cancer driver mutations. We find using alkaline comet assay, single-molecule DNA fiber assays, and immunofluorescence microscopy of DNA repair foci that H. pylori induces transcription-dependent DNA damage in actively replicating, Leucine-rich-repeat containing G-Protein-Coupled Receptor 5 (Lgr5)-positive antral stem and progenitor cells and their Troy-positive corpus counterparts, but not in other gastric epithelial lineages. Infection-dependent DNA damage is aggravated by Apc inactivation, but not by Trp53 or Smad4 loss, or Erbb2 overexpression. Our data suggest that H. pylori induces DNA damage in stem and progenitor cells, especially in settings of hyperproliferation due to constitutively active Wnt signaling.
- Published
- 2023
- Full Text
- View/download PDF
137. Reactivation of embryonic genetic programs in tissue regeneration and disease.
- Author
-
Fazilaty H and Basler K
- Subjects
- Humans, Cell Plasticity, Disease Progression, Embryonic Development genetics
- Abstract
Embryonic genetic programs are reactivated in response to various types of tissue damage, providing cell plasticity for tissue regeneration or disease progression. In acute conditions, these programs remedy the damage and then halt to allow a return to homeostasis. In chronic situations, including inflammatory diseases, fibrosis and cancer, prolonged activation of embryonic programs leads to disease progression and tissue deterioration. Induction of progenitor identity and cell plasticity, for example, epithelial-mesenchymal plasticity, are critical outcomes of reactivated embryonic programs. In this Review, we describe molecular players governing reactivated embryonic genetic programs, their role during disease progression, their similarities and differences and lineage reversion in pathology and discuss associated therapeutics and drug-resistance mechanisms across many organs. We also discuss the diversity of reactivated programs in different disease contexts. A comprehensive overview of commonalities between development and disease will provide better understanding of the biology and therapeutic strategies., (© 2023. Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
138. The diverse nature of intestinal fibroblasts in development, homeostasis, and disease.
- Author
-
Brügger MD and Basler K
- Subjects
- Humans, Mice, Animals, Homeostasis, Fibroblasts metabolism, Intestines
- Abstract
Only in recent years have we begun to appreciate the involvement of fibroblasts in intestinal development, tissue homeostasis, and disease. These insights followed the advent of single-cell transcriptomics that allowed researchers to explore the heterogeneity of intestinal fibroblasts in unprecedented detail. Since researchers often defined cell types and their associated function based on the biological process they studied, there are a plethora of partially overlapping markers for different intestinal fibroblast populations. This ambiguity complicates putting different research findings into context. Here, we provide a census on the function and identity of intestinal fibroblasts in mouse and human. We propose a simplified framework consisting of three colonic and four small intestinal fibroblast populations to aid navigating the diversity of intestinal fibroblasts., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
139. Terminal differentiation of villus tip enterocytes is governed by distinct Tgfβ superfamily members.
- Author
-
Berková L, Fazilaty H, Yang Q, Kubovčiak J, Stastna M, Hrckulak D, Vojtechova M, Dalessi T, Brügger MD, Hausmann G, Liberali P, Korinek V, Basler K, and Valenta T
- Subjects
- Ligands, Transforming Growth Factor beta metabolism, Bone Morphogenetic Proteins metabolism, Cell Differentiation, Enterocytes metabolism, Intestinal Mucosa metabolism
- Abstract
The protective and absorptive functions of the intestinal epithelium rely on differentiated enterocytes in the villi. The differentiation of enterocytes is orchestrated by sub-epithelial mesenchymal cells producing distinct ligands along the villus axis, in particular Bmps and Tgfβ. Here, we show that individual Bmp ligands and Tgfβ drive distinct enterocytic programs specific to villus zonation. Bmp4 is expressed from the centre to the upper part of the villus and activates preferentially genes connected to lipid uptake and metabolism. In contrast, Bmp2 is produced by villus tip mesenchymal cells and it influences the adhesive properties of villus tip epithelial cells and the expression of immunomodulators. Additionally, Tgfβ induces epithelial gene expression programs similar to those triggered by Bmp2. Bmp2-driven villus tip program is activated by a canonical Bmp receptor type I/Smad-dependent mechanism. Finally, we establish an organoid cultivation system that enriches villus tip enterocytes and thereby better mimics the cellular composition of the intestinal epithelium. Our data suggest that not only a Bmp gradient but also the activity of individual Bmp drives specific enterocytic programs., (© 2023 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
140. Wnt/β-Catenin Signaling Pathway Is Necessary for the Specification but Not the Maintenance of the Mouse Retinal Pigment Epithelium.
- Author
-
Kim JM, Min KW, Kim YJ, Smits R, Basler K, and Kim JW
- Subjects
- Mice, Animals, Cell Differentiation, beta Catenin genetics, beta Catenin metabolism, Gene Expression Regulation, Neurons metabolism, Retinal Pigment Epithelium metabolism, Wnt Signaling Pathway genetics
- Abstract
β-Catenin (Ctnnb1) has been shown to play critical roles in the development and maintenance of epithelial cells, including the retinal pigment epithelium (RPE). Ctnnb1 is not only a component of intercellular junctions in the epithelium, it also functions as a transcriptional regulator in the Wnt signaling pathway. To identify which of its functional modalities is critically involved in mouse RPE development and maintenance, we varied Ctnnb1 gene content and activity in mouse RPE lineage cells and tested their impacts on mouse eye development. We found that a Ctnnb1 double mutant (Ctnnb1
dm ), which exhibits impaired transcriptional activity, could not replace Ctnnb1 in the RPE, whereas Ctnnb1Y654E , which has reduced affinity for the junctions, could do so. Expression of the constitutively active Ctnnb1∆ex3 mutant also suppressed the development of RPE, instead facilitating a ciliary cell fate. However, the post-mitotic or mature RPE was insensitive to the loss, inactivation, or constitutive activation of Ctnnb1. Collectively, our results suggest that Ctnnb1 should be maintained within an optimal range to specify RPE through transcriptional regulation of Wnt target genes in the optic neuroepithelium.- Published
- 2023
- Full Text
- View/download PDF
141. Active eosinophils regulate host defence and immune responses in colitis.
- Author
-
Gurtner A, Borrelli C, Gonzalez-Perez I, Bach K, Acar IE, Núñez NG, Crepaz D, Handler K, Vu VP, Lafzi A, Stirm K, Raju D, Gschwend J, Basler K, Schneider C, Slack E, Valenta T, Becher B, Krebs P, Moor AE, and Arnold IC
- Subjects
- Animals, Humans, Mice, Inflammatory Bowel Diseases immunology, Single-Cell Gene Expression Analysis, Transcriptome, Proteome, Interleukin-33, Interferon-gamma, T-Lymphocytes, B7-1 Antigen metabolism, Colitis immunology, Colitis pathology, Eosinophils classification, Eosinophils cytology, Eosinophils immunology, Eosinophils metabolism, Immunity, Intestines immunology, Intestines pathology
- Abstract
In the past decade, single-cell transcriptomics has helped to uncover new cell types and states and led to the construction of a cellular compendium of health and disease. Despite this progress, some difficult-to-sequence cells remain absent from tissue atlases. Eosinophils-elusive granulocytes that are implicated in a plethora of human pathologies
1-5 -are among these uncharted cell types. The heterogeneity of eosinophils and the gene programs that underpin their pleiotropic functions remain poorly understood. Here we provide a comprehensive single-cell transcriptomic profiling of mouse eosinophils. We identify an active and a basal population of intestinal eosinophils, which differ in their transcriptome, surface proteome and spatial localization. By means of a genome-wide CRISPR inhibition screen and functional assays, we reveal a mechanism by which interleukin-33 (IL-33) and interferon-γ (IFNγ) induce the accumulation of active eosinophils in the inflamed colon. Active eosinophils are endowed with bactericidal and T cell regulatory activity, and express the co-stimulatory molecules CD80 and PD-L1. Notably, active eosinophils are enriched in the lamina propria of a small cohort of patients with inflammatory bowel disease, and are closely associated with CD4+ T cells. Our findings provide insights into the biology of eosinophils and highlight the crucial contribution of this cell type to intestinal homeostasis, immune regulation and host defence. Furthermore, we lay a framework for the characterization of eosinophils in human gastrointestinal diseases., (© 2022. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
142. Glycoprotein (GP)96 Is Essential for Maintaining Intestinal Epithelial Architecture by Supporting Its Self-Renewal Capacity.
- Author
-
Häfliger J, Schwarzfischer M, Atrott K, Stanzel C, Morsy Y, Wawrzyniak M, Lang S, Valenta T, Basler K, Rogler G, Scharl M, and Spalinger MR
- Subjects
- Animals, Mice, Cell Proliferation, Glycoproteins metabolism, Wnt Signaling Pathway genetics, Epithelial Cells metabolism, Intestines cytology, Membrane Glycoproteins metabolism
- Abstract
Background & Aims: Glycoprotein (GP)96 is an endoplasmic reticulum-resident master chaperone for cell surface receptors including the Wnt co-receptors low-density lipoprotein-receptor-related protein 5/6. Intestinal epithelial cell (IEC)-specific deletion of Gp96 is embryonically lethal. However, the role of GP96 in adult intestinal tissue and especially within the intestinal stem cell (ISC) niche is unknown. Here, we investigated how GP96 loss interferes with intestinal homeostasis by compromising viability, proliferation, and differentiation of IECs., Methods: Tamoxifen was used to induce Cre-mediated deletion of Gp96 in GP96-Villin
creERT2 (Cre recombinase-Estrogen-Receptor Transgene 2) mice and intestinal organoids. With H&E and immunofluorescence staining we assessed alterations in intestinal morphology and the presence and localization of IEC types. Real-time polymerase chain reaction and Western blot analysis were performed to explore the molecular mechanisms underlying the severe phenotype of Gp96 KO mice and organoids., Results: IEC-specific deletion of Gp96 in adult mice resulted in a rapid degeneration of the stem cell niche, followed by complete eradication of the epithelial layer and death within a few days. These effects were owing to severe defects in ISC renewal and premature ISC differentiation, which resulted from defective Wnt and Notch signaling. Furthermore, depletion of GP96 led to massive induction of endoplasmic reticulum stress. Although effects on ISC renewal and adequate differentiation were partly reversed upon activation of Wnt/Notch signaling, viability could not be restored, indicating that reduced viability was mediated by other mechanisms., Conclusions: Our work shows that GP96 plays a fundamental role in regulating ISC fate and epithelial regeneration and therefore is indispensable for maintaining intestinal epithelial homeostasis., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
143. GATA3 mediates nonclassical β-catenin signaling in skeletal cell fate determination and ectopic chondrogenesis.
- Author
-
Maruyama T, Hasegawa D, Valenta T, Haigh J, Bouchard M, Basler K, and Hsu W
- Subjects
- Animals, Mice, Chondrogenesis genetics, Cell Differentiation genetics, Wnt Signaling Pathway, GATA3 Transcription Factor genetics, beta Catenin genetics, Musculoskeletal System
- Abstract
Skeletal precursors are mesenchymal in origin and can give rise to distinct sublineages. Their lineage commitment is modulated by various signaling pathways. The importance of Wnt signaling in skeletal lineage commitment has been implicated by the study of β-catenin-deficient mouse models. Ectopic chondrogenesis caused by the loss of β-catenin leads to a long-standing belief in canonical Wnt signaling that determines skeletal cell fate. As β-catenin has other functions, it remains unclear whether skeletogenic lineage commitment is solely orchestrated by canonical Wnt signaling. The study of the Wnt secretion regulator Gpr177/Wntless also raises concerns about current knowledge. Here, we show that skeletal cell fate is determined by β-catenin but independent of LEF/TCF transcription. Genomic and bioinformatic analyses further identify GATA3 as a mediator for the alternative signaling effects. GATA3 alone is sufficient to promote ectopic cartilage formation, demonstrating its essential role in mediating nonclassical β-catenin signaling in skeletogenic lineage specification.
- Published
- 2022
- Full Text
- View/download PDF
144. Epithelial Wnt secretion drives the progression of inflammation-induced colon carcinoma in murine model.
- Author
-
Degirmenci B, Dincer C, Demirel HC, Berkova L, Moor AE, Kahraman A, Hausmann G, Aguet M, Tuncbag N, Valenta T, and Basler K
- Abstract
Colon cancer is initiated by stem cells that escape the strict control. This process is often driven through aberrant activation of Wnt signaling by mutations in components acting downstream of the receptor complex that unfetter tumor cells from the need for Wnts. Here we describe a class of colon cancer that does not depend on mutated core components of the Wnt pathway. Genetically blocking Wnt secretion from epithelial cells of such tumors results in apoptosis, reduced expression of colon cancer markers, followed by enhanced tumor differentiation. In contrast to the normal colonic epithelium, such tumor cells autosecrete Wnts to maintain their uncontrolled proliferative behavior. In humans, we determined certain cases of colon cancers in which the Wnt pathway is hyperactive, but not through mutations in its core components. Our findings illuminate the path in therapy to find further subtypes of Wnt-dependent colon cancer that might be responsive to Wnt secretion inhibitors., Competing Interests: The authors declare no competing of interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
145. Only the Co-Transcriptional Activity of β-Catenin Is Required for the Local Regulatory Effects in Hypertrophic Chondrocytes on Developmental Bone Modeling.
- Author
-
Wolff LI, Houben A, Fabritius C, Angus-Hill M, Basler K, and Hartmann C
- Subjects
- Animals, Cell Differentiation, Mice, Osteoclasts metabolism, TCF Transcription Factors, Wnt Signaling Pathway, Chondrocytes metabolism, beta Catenin genetics, beta Catenin metabolism
- Abstract
In hypertrophic chondrocytes, β-catenin has two roles. First, it locally suppresses the differentiation of osteoclasts at the chondro-osseous junction by maintaining the pro-osteoclastic factor receptor activator of NF-κB ligand (RANKL) at low levels. Second, it promotes the differentiation of osteoblast-precursors from chondrocytes. Yet, β-catenin is a dual-function protein, which can either participate in cell-cell adherens junctions or serve as a transcriptional co-activator in canonical Wnt signaling interacting with T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcription factors. Hence, whenever studying tissue-specific requirements of β-catenin using a conventional conditional knockout approach, the functional mechanisms underlying the defects in the conditional mutants remain ambiguous. To decipher mechanistically which of the two molecular functions of β-catenin is required in hypertrophic chondrocytes, we used different approaches. We analyzed the long bones of newborn mice carrying either the null-alleles of Lef1 or Tcf7, or mice in which Tcf7l2 was conditionally deleted in the hypertrophic chondrocytes, as well as double mutants for Lef1 and Tcf7l2, and Tcf7 and Tcf7l2. Furthermore, we analyzed Ctnnb1 mutant newborns expressing a signaling-defective allele that retains the cell adhesion function in hypertrophic chondrocytes. None of the analyzed Tcf/Lef single or double mutants recapitulated the previously published phenotype upon loss of β-catenin in hypertrophic chondrocytes. However, using this particular Ctnnb1 allele, maintaining cell adhesion function, we show that it is the co-transcriptional activity of β-catenin, which is required in hypertrophic chondrocytes to suppress osteoclastogenesis and to promote chondrocyte-derived osteoblast differentiation. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)., (© 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).)
- Published
- 2021
- Full Text
- View/download PDF
146. The interactions of Bcl9/Bcl9L with β-catenin and Pygopus promote breast cancer growth, invasion, and metastasis.
- Author
-
Vafaizadeh V, Buechel D, Rubinstein N, Kalathur RKR, Bazzani L, Saxena M, Valenta T, Hausmann G, Cantù C, Basler K, and Christofori G
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, DNA-Binding Proteins genetics, Disease Progression, Epithelial-Mesenchymal Transition, Female, Humans, Mice, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Transcription Factors genetics, Wnt Signaling Pathway, beta Catenin genetics, Breast Neoplasms pathology, DNA-Binding Proteins metabolism, Intracellular Signaling Peptides and Proteins metabolism, Transcription Factors metabolism, beta Catenin metabolism
- Abstract
Canonical Wnt/β-catenin signaling is an established regulator of cellular state and its critical contributions to tumor initiation, malignant tumor progression and metastasis formation have been demonstrated in various cancer types. Here, we investigated how the binding of β-catenin to the transcriptional coactivators B-cell CLL/lymphoma 9 (Bcl9) and Bcl9-Like (Bcl9L) affected mammary gland carcinogenesis in the MMTV-PyMT transgenic mouse model of metastatic breast cancer. Conditional knockout of both Bcl9 and Bcl9L resulted into tumor cell death. In contrast, disrupting the interaction of Bcl9/Bcl9L with β-catenin, either by deletion of their HD2 domains or by a point mutation in the N-terminal domain of β-catenin (D164A), diminished primary tumor growth and tumor cell proliferation and reduced tumor cell invasion and lung metastasis. In comparison, the disruption of HD1 domain-mediated binding of Bcl9/Bcl9L to Pygopus had only moderate effects. Interestingly, interfering with the β-catenin-Bcl9/Bcl9L-Pygo chain of adapters only partially impaired the transcriptional response of mammary tumor cells to Wnt3a and TGFβ treatments. Together, the results indicate that Bcl9/Bcl9L modulate but are not critically required for canonical Wnt signaling in its contribution to breast cancer growth and malignant progression, a notion consistent with the "just-right" hypothesis of Wnt-driven tumor progression., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
147. TCF/LEF regulation of the topologically associated domain ADI promotes mESCs to exit the pluripotent ground state.
- Author
-
Doumpas N, Söderholm S, Narula S, Moreira S, Doble BW, Cantù C, and Basler K
- Subjects
- Animals, Benzamides pharmacology, Culture Media chemistry, Culture Media pharmacology, DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, DNA (Cytosine-5-)-Methyltransferases genetics, DNA (Cytosine-5-)-Methyltransferases metabolism, Diphenylamine analogs & derivatives, Diphenylamine pharmacology, Down-Regulation drug effects, Gene Editing, Hepatocyte Nuclear Factor 1-alpha deficiency, Hepatocyte Nuclear Factor 1-alpha metabolism, Inducible T-Cell Co-Stimulator Ligand antagonists & inhibitors, Inducible T-Cell Co-Stimulator Ligand genetics, Inducible T-Cell Co-Stimulator Ligand metabolism, Lymphoid Enhancer-Binding Factor 1 deficiency, Lymphoid Enhancer-Binding Factor 1 metabolism, Mice, Mouse Embryonic Stem Cells cytology, Mouse Embryonic Stem Cells metabolism, Octamer Transcription Factor-3 genetics, Octamer Transcription Factor-3 metabolism, Pyridines pharmacology, Pyrimidines pharmacology, RNA Interference, RNA, Small Interfering metabolism, Transcription Factor 7-Like 1 Protein deficiency, Transcription Factor 7-Like 1 Protein metabolism, Transcription Factor 7-Like 2 Protein deficiency, Transcription Factor 7-Like 2 Protein metabolism, Transcription Factors antagonists & inhibitors, Transcription Factors genetics, Transcription Factors metabolism, beta Catenin deficiency, beta Catenin genetics, AIRE Protein, Cell Self Renewal drug effects, Hepatocyte Nuclear Factor 1-alpha genetics, Lymphoid Enhancer-Binding Factor 1 genetics, Transcription Factor 7-Like 1 Protein genetics, Transcription Factor 7-Like 2 Protein genetics
- Abstract
Mouse embryonic stem cells (mESCs) can be maintained in vitro in defined N2B27 medium supplemented with two chemical inhibitors for GSK3 and MEK (2i) and the cytokine leukemia inhibitory factor (LIF), which act synergistically to promote self-renewal and pluripotency. Here, we find that genetic deletion of the four genes encoding the TCF/LEF transcription factors confers mESCs with the ability to self-renew in N2B27 medium alone. TCF/LEF quadruple knockout (qKO) mESCs display dysregulation of several genes, including Aire, Dnmt3l, and IcosL, located adjacent to each other within a topologically associated domain (TAD). Aire, Dnmt3l, and IcosL appear to be regulated by TCF/LEF in a β-catenin independent manner. Moreover, downregulation of Aire and Dnmt3l in wild-type mESCs mimics the loss of TCF/LEF and increases mESC survival in the absence of 2iL. Hence, this study identifies TCF/LEF effectors that mediate exit from the pluripotent state., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
148. Parsing β-catenin's cell adhesion and Wnt signaling functions in malignant mammary tumor progression.
- Author
-
Buechel D, Sugiyama N, Rubinstein N, Saxena M, Kalathur RKR, Lüönd F, Vafaizadeh V, Valenta T, Hausmann G, Cantù C, Basler K, and Christofori G
- Subjects
- Animals, Apoptosis, Cell Cycle, Cell Movement, Epithelial-Mesenchymal Transition drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Mammary Neoplasms, Animal genetics, Mice, Mice, Transgenic, Neoplasm Invasiveness, Neoplasm Metastasis, Transcriptome, Transforming Growth Factor beta pharmacology, Wnt3A Protein genetics, beta Catenin genetics, Cell Adhesion physiology, Mammary Neoplasms, Animal metabolism, Signal Transduction physiology, Wnt3A Protein metabolism, beta Catenin metabolism
- Abstract
During malignant progression, epithelial cancer cells dissolve their cell-cell adhesion and gain invasive features. By virtue of its dual function, β-catenin contributes to cadherin-mediated cell-cell adhesion, and it determines the transcriptional output of Wnt signaling: via its N terminus, it recruits the signaling coactivators Bcl9 and Pygopus, and via the C terminus, it interacts with the general transcriptional machinery. This duality confounds the simple loss-of-function analysis of Wnt signaling in cancer progression. In many cancer types including breast cancer, the functional contribution of β-catenin's transcriptional activities, as compared to its adhesion functions, to tumor progression has remained elusive. Employing the mouse mammary tumor virus (MMTV)-PyMT mouse model of metastatic breast cancer, we compared the complete elimination of β-catenin with the specific ablation of its signaling outputs in mammary tumor cells. Notably, the complete lack of β-catenin resulted in massive apoptosis of mammary tumor cells. In contrast, the loss of β-catenin's transcriptional activity resulted in a reduction of primary tumor growth, tumor invasion, and metastasis formation in vivo. These phenotypic changes were reflected by stalled cell cycle progression and diminished epithelial-mesenchymal transition (EMT) and cell migration of breast cancer cells in vitro. Transcriptome analysis revealed subsets of genes which were specifically regulated by β-catenin's transcriptional activities upon stimulation with Wnt3a or during TGF-β-induced EMT. Our results uncouple the signaling from the adhesion function of β-catenin and underline the importance of Wnt/β-catenin-dependent transcription in malignant tumor progression of breast cancer., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
149. Epigenetic control of melanoma cell invasiveness by the stem cell factor SALL4.
- Author
-
Diener J, Baggiolini A, Pernebrink M, Dalcher D, Lerra L, Cheng PF, Varum S, Häusel J, Stierli S, Treier M, Studer L, Basler K, Levesque MP, Dummer R, Santoro R, Cantù C, and Sommer L
- Subjects
- Acetylation, Animals, Base Sequence, Carcinogenesis genetics, Carcinogenesis pathology, Cell Adhesion genetics, Cell Line, Tumor, Cell Lineage, Cell Proliferation, DNA-Binding Proteins metabolism, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Histone Deacetylase 2 metabolism, Histones metabolism, Humans, Melanocytes metabolism, Melanocytes pathology, Mice, Nude, Mice, Transgenic, Neoplasm Invasiveness, Neoplasm Micrometastasis, Protein Binding, Tumor Burden, Mice, Epigenesis, Genetic, Melanoma genetics, Melanoma pathology, Skin Neoplasms genetics, Skin Neoplasms pathology, Stem Cell Factor metabolism, Transcription Factors metabolism
- Abstract
Melanoma cells rely on developmental programs during tumor initiation and progression. Here we show that the embryonic stem cell (ESC) factor Sall4 is re-expressed in the Tyr::Nras
Q61K ; Cdkn2a-/- melanoma model and that its expression is necessary for primary melanoma formation. Surprisingly, while Sall4 loss prevents tumor formation, it promotes micrometastases to distant organs in this melanoma-prone mouse model. Transcriptional profiling and in vitro assays using human melanoma cells demonstrate that SALL4 loss induces a phenotype switch and the acquisition of an invasive phenotype. We show that SALL4 negatively regulates invasiveness through interaction with the histone deacetylase (HDAC) 2 and direct co-binding to a set of invasiveness genes. Consequently, SALL4 knock down, as well as HDAC inhibition, promote the expression of an invasive signature, while inhibition of histone acetylation partially reverts the invasiveness program induced by SALL4 loss. Thus, SALL4 appears to regulate phenotype switching in melanoma through an HDAC2-mediated mechanism., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
150. Tracing colonic embryonic transcriptional profiles and their reactivation upon intestinal damage.
- Author
-
Fazilaty H, Brügger MD, Valenta T, Szczerba BM, Berkova L, Doumpas N, Hausmann G, Scharl M, and Basler K
- Subjects
- Animals, Cell Differentiation, Colitis genetics, Disease Models, Animal, Embryo, Mammalian metabolism, Gene Expression Regulation, Developmental, Humans, Inflammatory Bowel Diseases genetics, Inflammatory Bowel Diseases pathology, Intestinal Mucosa embryology, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Mesoderm embryology, Mice, Inbred C57BL, Single-Cell Analysis, Mice, Colon embryology, Colon pathology, Gene Expression Profiling
- Abstract
We lack a holistic understanding of the genetic programs orchestrating embryonic colon morphogenesis and governing damage response in the adult. A window into these programs is the transcriptomes of the epithelial and mesenchymal cell populations in the colon. Performing unbiased single-cell transcriptomic analyses of the developing mouse colon at different embryonic stages (embryonic day 14.5 [E14.5], E15.5, and E18.5), we capture cellular and molecular profiles of the stages before, during, and after the appearance of crypt structures, as well as in a model of adult colitis. The data suggest most adult lineages are established by E18.5. We find embryonic-specific gene expression profiles and cell populations that reappear in response to tissue damage. Comparison of the datasets from mice and human colitis suggests the processes are conserved. In this study, we provide a comprehensive single-cell atlas of the developing mouse colon and evidence for the reactivation of embryonic genes in disease., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.