1
|
Ragab EM, Gamal DME, El-Najjar FF, Elkomy HA, Ragab MA, Elantary MA, Basyouni OM, Moustafa SM, El-Naggar SA, Elsherbiny AS. New insights into Notch signaling as a crucial pathway of pancreatic cancer stem cell behavior by chrysin-polylactic acid-based nanocomposite. Discov Oncol 2025; 16:107. [PMID: 39891818 PMCID: PMC11787125 DOI: 10.1007/s12672-025-01846-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Pancreatic cancer is an extremely deadly illness for which there are few reliable treatments. Recent research indicates that malignant tumors are highly variable and consist of a tiny subset of unique cancer cells, known as cancer stem cells (CSCs), which are responsible for the beginning and spread of tumors. These cells are typically identified by the expression of specific cell surface markers. A population of pancreatic cancer stem cells with aberrantly active developmental signaling pathways has been identified in recent studies of human pancreatic tumors. Among these Notch signaling pathway has been identified as a key regulator of CSCs self-renewal, making it an attractive target for therapeutic intervention. Chrysin-loaded polylactic acid (PLA) as polymeric nanoparticles systems have been growing interest in using as platforms for improved drug delivery. This review aims to explore innovative strategies for targeted therapy and optimized drug delivery in pancreatic CSCs by manipulating the Notch pathway and leveraging PLA-based drug delivery systems. Furthermore, we will assess the capability of PLA nanoparticles to enhance the bioavailability and effectiveness of gemcitabine in pancreatic cancer cells. The insights gained from this review have the potential to contribute to the development of novel treatment approaches that combine targeted therapy with advanced drug delivery utilizing biodegradable polymeric nanoparticles.
Collapse
Affiliation(s)
- Eman M Ragab
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Fares F El-Najjar
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hager A Elkomy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A Ragab
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mariam A Elantary
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Omar M Basyouni
- Chemistry/Zoology Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sherif M Moustafa
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa A El-Naggar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Abeer S Elsherbiny
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
2
|
Nair S, Baker NE. Extramacrochaetae regulates Notch signaling in the Drosophila eye through non-apoptotic caspase activity. eLife 2024; 12:RP91988. [PMID: 39564985 PMCID: PMC11578588 DOI: 10.7554/elife.91988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Abstract
Many cell fate decisions are determined transcriptionally. Accordingly, some fate specification is prevented by Inhibitor of DNA-binding (Id) proteins that interfere with DNA binding by master regulatory transcription factors. We show that the Drosophila Id protein Extra macrochaetae (Emc) also affects developmental decisions by regulating caspase activity. Emc, which prevents proneural bHLH transcription factors from specifying neural cell fate, also prevents homodimerization of another bHLH protein, Daughterless (Da), and thereby maintains expression of the Death-Associated Inhibitor of Apoptosis (diap1) gene. Accordingly, we found that multiple effects of emc mutations on cell growth and on eye development were all caused by activation of caspases. These effects included acceleration of the morphogenetic furrow, failure of R7 photoreceptor cell specification, and delayed differentiation of non-neuronal cone cells. Within emc mutant clones, Notch signaling was elevated in the morphogenetic furrow, increasing morphogenetic furrow speed. This was associated with caspase-dependent increase in levels of Delta protein, the transmembrane ligand for Notch. Posterior to the morphogenetic furrow, elevated Delta cis-inhibited Notch signaling that was required for R7 specification and cone cell differentiation. Growth inhibition of emc mutant clones in wing imaginal discs also depended on caspases. Thus, emc mutations reveal the importance of restraining caspase activity even in non-apoptotic cells to prevent abnormal development, in the Drosophila eye through effects on Notch signaling.
Collapse
Affiliation(s)
- Sudershana Nair
- Department of Genetics, Albert Einstein College of MedicineBronxUnited States
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of MedicineBronxUnited States
- Department of Developmental and Molecular Biology, Albert Einstein College of MedicineBronxUnited States
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
3
|
Nair S, Baker NE. Extramacrochaetae regulates Notch signaling in the Drosophila eye through non-apoptotic caspase activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.04.560841. [PMID: 39131389 PMCID: PMC11312471 DOI: 10.1101/2023.10.04.560841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Many cell fate decisions are determined transcriptionally. Accordingly, some fate specification is prevented by Inhibitor of DNA binding (Id) proteins that interfere with DNA binding by master regulatory transcription factors. We show that the Drosophila Id protein Extra macrochaetae (Emc) also affects developmental decisions by regulating caspase activity. Emc, which prevents proneural bHLH transcription factors from specifying neural cell fate, also prevents homodimerization of another bHLH protein, Daughterless (Da), and thereby maintains expression of the Death-Associated Inhibitor of Apoptosis (diap1) gene. Accordingly, we found that multiple effects of emc mutations on cell growth and on eye development were all caused by activation of caspases. These effects included acceleration of the morphogenetic furrow, failure of R7 photoreceptor cell specification, and delayed differentiation of non-neuronal cone cells. Within emc mutant clones, Notch signaling was elevated in the morphogenetic furrow, increasing morphogenetic furrow speed. This was associated with caspase-dependent increase in levels of Delta protein, the transmembrane ligand for Notch. Posterior to the morphogenetic furrow, elevated Delta cis-inhibited Notch signaling that was required for R7 specification and cone cell differentiation. Growth inhibition of emc mutant clones in wing imaginal discs also depended on caspases. Thus, emc mutations reveal the importance of restraining caspase activity even in non-apoptotic cells to prevent abnormal development, in the Drosophila eye through effects on Notch signaling.
Collapse
Affiliation(s)
- Sudershana Nair
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Present address: Department of and Physiology, NYU School of Medicine, 435 East 30 St, New York, NY
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Present address: Department of Microbiology and Molecular Genetics, University of California, Irvine, 2011 Biological Sciences 3, Irvine, CA 92697-2300
| |
Collapse
|
4
|
Cao R, Gozlan O, Airich A, Tveriakhina L, Zhou H, Jiang H, Cole PA, Aster JC, Klein T, Sprinzak D, Blacklow SC. Structural requirements for activity of Mind bomb1 in Notch signaling. Structure 2024; 32:1667-1676.e5. [PMID: 39121852 DOI: 10.1016/j.str.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/13/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Abstract
Mind bomb 1 (MIB1) is a RING E3 ligase that ubiquitinates Notch ligands, a necessary step for induction of Notch signaling. The structural basis for binding of the JAG1 ligand by the N-terminal region of MIB1 is known, yet how the ankyrin (ANK) and RING domains of MIB1 cooperate to catalyze ubiquitin transfer from E2∼Ub to Notch ligands remains unclear. Here, we show that the third RING domain and adjacent coiled coil region (ccRING3) drive MIB1 dimerization and that MIB1 ubiquitin transfer activity relies solely on ccRING3. We report X-ray crystal structures of a UbcH5B-ccRING3 complex and the ANK domain. Directly tethering the MIB1 N-terminal region to ccRING3 forms a minimal MIB1 protein sufficient to induce a Notch response in receiver cells and rescue mib knockout phenotypes in flies. Together, these studies define the functional elements of an E3 ligase needed for ligands to induce a Notch signaling response.
Collapse
Affiliation(s)
- Ruili Cao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Oren Gozlan
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel
| | - Alina Airich
- Institute of Genetics, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Lena Tveriakhina
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Haixia Zhou
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hanjie Jiang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Philip A Cole
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Jon C Aster
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - David Sprinzak
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
5
|
Pinot M, Le Borgne R. Spatio-Temporal Regulation of Notch Activation in Asymmetrically Dividing Sensory Organ Precursor Cells in Drosophila melanogaster Epithelium. Cells 2024; 13:1133. [PMID: 38994985 PMCID: PMC11240559 DOI: 10.3390/cells13131133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The Notch communication pathway, discovered in Drosophila over 100 years ago, regulates a wide range of intra-lineage decisions in metazoans. The division of the Drosophila mechanosensory organ precursor is the archetype of asymmetric cell division in which differential Notch activation takes place at cytokinesis. Here, we review the molecular mechanisms by which epithelial cell polarity, cell cycle and intracellular trafficking participate in controlling the directionality, subcellular localization and temporality of mechanosensitive Notch receptor activation in cytokinesis.
Collapse
Affiliation(s)
| | - Roland Le Borgne
- Univ Rennes, Centre National de la Recherche Scientifique UMR 6290, IGDR (Institut de Génétique et Développement de Rennes), F-35000 Rennes, France
| |
Collapse
|
6
|
Tveriakhina L, Scanavachi G, Egan ED, Da Cunha Correia RB, Martin AP, Rogers JM, Yodh JS, Aster JC, Kirchhausen T, Blacklow SC. Temporal dynamics and stoichiometry in human Notch signaling from Notch synaptic complex formation to nuclear entry of the Notch intracellular domain. Dev Cell 2024; 59:1425-1438.e8. [PMID: 38574735 DOI: 10.1016/j.devcel.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
Mammalian Notch signaling occurs when the binding of Delta or Jagged to Notch stimulates the proteolytic release of the Notch intracellular domain (NICD), which enters the nucleus to control target gene expression. To determine the temporal dynamics of events associated with Notch signaling under native conditions, we fluorescently tagged Notch and Delta at their endogenous genomic loci and visualized them upon pairing of receiver (Notch) and sender (Delta) cells as a function of time after cell contact. At contact sites, Notch and Delta immediately accumulated at 1:1 stoichiometry in synapses, which resolved by 15-20 min after contact. Synapse formation preceded the entrance of the Notch extracellular domain into the sender cell and accumulation of NICD in the nucleus of the receiver cell, which approached a maximum after ∼45 min and was prevented by chemical and genetic inhibitors of signaling. These findings directly link Notch-Delta synapse dynamics to NICD production with spatiotemporal precision.
Collapse
Affiliation(s)
- Lena Tveriakhina
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gustavo Scanavachi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Emily D Egan
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ricardo Bango Da Cunha Correia
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Alexandre P Martin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Julia M Rogers
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy S Yodh
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
7
|
Cao R, Gozlan O, Tveriakhina L, Zhou H, Jiang H, Cole PA, Aster JC, Sprinzak D, Blacklow SC. Structural Requirements for Activity of Mind bomb1 in Notch Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582834. [PMID: 38464278 PMCID: PMC10925295 DOI: 10.1101/2024.03.01.582834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Mind bomb 1 (MIB1) is a RING E3 ligase that ubiquitinates Notch ligands, a necessary step for induction of Notch signaling. The structural basis for binding of the JAG1 ligand by the N-terminal region of MIB1 is known, yet how the ankyrin (ANK) and RING domains of MIB1 cooperate to catalyze ubiquitin transfer from E2~Ub to Notch ligands remains unclear. Here, we show that the third RING domain and adjacent coiled coil region of MIB1 (ccRING3) drives MIB1 dimerization and that ubiquitin transfer activity of MIB1 relies solely on RING3. We report x-ray crystal structures of a UbcH5B-ccRING3 complex as a fusion protein and of the ANK region. Directly tethering the N-terminal region to ccRING3 forms a minimal MIB1 protein, which is sufficient to induce a Notch response in receiver cells. Together, these studies define the functional elements of an E3 ligase needed for ligands to induce a Notch signaling response.
Collapse
Affiliation(s)
- Ruili Cao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Oren Gozlan
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Tveriakhina
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Haixia Zhou
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hanjie Jiang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Philip A Cole
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Jon C Aster
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - David Sprinzak
- George S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry, and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Lead contact
| |
Collapse
|
8
|
Kalodimou K, Stapountzi M, Vüllings N, Seib E, Klein T, Delidakis C. Separable Roles for Neur and Ubiquitin in Delta Signalling in the Drosophila CNS Lineages. Cells 2023; 12:2833. [PMID: 38132160 PMCID: PMC10741450 DOI: 10.3390/cells12242833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
The execution of a Notch signal at the plasma membrane relies on the mechanical force exerted onto Notch by its ligand. It has been appreciated that the DSL ligands need to collaborate with a ubiquitin (Ub) ligase, either Neuralized or Mindbomb1, in order to exert this pulling force, but the role of ubiquitylation per se is uncertain. Regarding the Delta-Neur pair, it is documented that neither the Neur catalytic domain nor the Delta intracellular lysines (putative Ub acceptors) are needed for activity. Here, we present a dissection of the Delta activity using the Delta-Notch-dependent expression of Hey in newborn Drosophila neurons as a sensitive in vivo assay. We show that the Delta-Neur interaction per se, rather than ubiquitylation, is needed for activity, pointing to the existence of a Delta-Neur signaling complex. The Neur catalytic domain, although not strictly needed, greatly improves Delta-Neur complex functionality when the Delta lysines are mutated, suggesting that the ubiquitylation of some component of the complex, other than Delta, can enhance signaling. Since Hey expression is sensitive to the perturbation of endocytosis, we propose that the Delta-Neur complex triggers a force-generating endocytosis event that activates Notch in the adjacent cell.
Collapse
Affiliation(s)
- Konstantina Kalodimou
- Department of Biology, University of Crete, 700 13 Heraklion, Greece;
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| | - Margarita Stapountzi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| | - Nicole Vüllings
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, 40225 Duesseldorf, Germany
| | - Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, 40225 Duesseldorf, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, 40225 Duesseldorf, Germany
| | - Christos Delidakis
- Department of Biology, University of Crete, 700 13 Heraklion, Greece;
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 700 13 Heraklion, Greece;
| |
Collapse
|
9
|
Troost T, Seib E, Airich A, Vüllings N, Necakov A, De Renzis S, Klein T. The meaning of ubiquitylation of the DSL ligand Delta for the development of Drosophila. BMC Biol 2023; 21:260. [PMID: 37974242 PMCID: PMC10655352 DOI: 10.1186/s12915-023-01759-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Ubiquitylation (ubi) of the intracellular domain of the Notch ligand Delta (Dl) by the E3 ligases Neuralized (Neur) and Mindbomb1 (Mib1) on lysines (Ks) is thought to be essential for the its signalling activity. Nevertheless, we have previously shown that DlK2R-HA, a Dl variant where all Ks in its intracellular domain (ICD) are replaced by the structurally similar arginine (R), still possess weak activity if over-expressed. This suggests that ubi is not absolutely required for Dl signalling. However, it is not known whether the residual activity of DlK2R-HA is an effect of over-expression and, if not, whether DlK2R can provide sufficient activity for the whole development of Drosophila. RESULTS To clarify these issues, we generated and analysed DlattP-DlK2R-HA, a knock-in allele into the Dl locus. Our analysis of this allele reveals that the sole presence of one copy of DlattP-DlK2R-HA can provide sufficient activity for completion of development. It further indicates that while ubi is required for the full activity of Dl in Mib1-dependent processes, it is not essential for Neur-controlled neural development. We identify three modes of Dl signalling that are either dependent or independent of ubi. Importantly, all modes depend on the presence of the endocytic adapter Epsin. During activation of Dl, direct binding of Epsin appears not to be an essential requirement. In addition, our analysis further reveals that the Ks are required to tune down the cis-inhibitory interaction of Dl with Notch. CONCLUSIONS Our results indicate that Dl can activate the Notch pathway without ubi of its ICD. It signals via three modes. Ubi is specifically required for the Mib1-dependent processes and the adjustment of cis-inhibition. In contrast to Mib1, Neur can efficiently activate Dl without ubi. Neur probably acts as an endocytic co-adapter in addition to its role as E3 ligase. Endocytosis, regulated in a ubi-dependent or ubi-independent manner is required for signalling and also suppression of cis-inhibition. The findings clarify the role of ubi of the ligands during Notch signalling.
Collapse
Affiliation(s)
- Tobias Troost
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Alina Airich
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Nicole Vüllings
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany
| | - Aleksandar Necakov
- Department of Biological Science, Brock University, 1030, Ontario, L2S3A1, Canada
| | - Stefano De Renzis
- European Molecular Biology Laboratory, Developmental Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, 40225, Duesseldorf, Germany.
| |
Collapse
|
10
|
Tveriakhina L, Scanavachi G, Egan ED, Correia RBDC, Martin AP, Rogers JM, Yodh JS, Aster JC, Kirchhausen T, Blacklow SC. Temporal Dynamics and Stoichiometry in Notch Signaling - from Notch Synaptic Complex Formation to NICD Nuclear Entry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559780. [PMID: 37808809 PMCID: PMC10557745 DOI: 10.1101/2023.09.27.559780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Mammalian Notch signaling occurs when binding of Delta or Jagged to Notch stimulates proteolytic release of the Notch intracellular domain (NICD), which enters the nucleus to regulate target gene expression. To determine the temporal dynamics of events associated with Notch signaling under native conditions, we fluorescently tagged Notch and Delta at their endogenous genomic loci and visualized them upon pairing of receiver (Notch) and sender (Delta) cells as a function of time after cell contact. At contact sites, Notch and Delta immediately accumulated at 1:1 stoichiometry in synapses, which resolved by 15-20 min after contact. Synapse formation preceded entrance of the Notch extracellular domain into the sender cell and accumulation of NICD in the nucleus of the receiver cell, which approached a maximum after ∼45 min and was prevented by chemical and genetic inhibitors of signaling. These findings directly link Notch-Delta synapse dynamics to NICD production with unprecedented spatiotemporal precision.
Collapse
|
11
|
Voutyraki C, Choromidis A, Meligkounaki A, Vlachopoulos NA, Theodorou V, Grammenoudi S, Athanasiadis E, Monticelli S, Giangrande A, Delidakis C, Zacharioudaki E. Growth deregulation and interaction with host hemocytes contribute to tumor progression in a Drosophila brain tumor model. Proc Natl Acad Sci U S A 2023; 120:e2221601120. [PMID: 37549261 PMCID: PMC10438840 DOI: 10.1073/pnas.2221601120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 08/09/2023] Open
Abstract
Tumors constantly interact with their microenvironment. Here, we present data on a Notch-induced neural stem cell (NSC) tumor in Drosophila, which can be immortalized by serial transplantation in adult hosts. This tumor arises in the larva by virtue of the ability of Notch to suppress early differentiation-promoting factors in NSC progeny. Guided by transcriptome data, we have addressed both tumor-intrinsic and microenvironment-specific factors and how they contribute to tumor growth and host demise. The growth promoting factors Myc, Imp, and Insulin receptor in the tumor cells are important for tumor expansion and killing of the host. From the host's side, hemocytes, professional phagocytic blood cells, are found associated with tumor cells. Phagocytic receptors, like NimC1, are needed in hemocytes to enable them to capture and engulf tumor cells, restricting their growth. In addition to their protective role, hemocytes may also increase the host's morbidity by their propensity to produce damaging extracellular reactive oxygen species.
Collapse
Affiliation(s)
- Chrysanthi Voutyraki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Alexandros Choromidis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Anastasia Meligkounaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Nikolaos Andreas Vlachopoulos
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Vasiliki Theodorou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| | - Sofia Grammenoudi
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672Athens, Greece
| | - Emmanouil Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, 11527Athens, Greece
- Medical Image and Signal Processing Laboratory, Department of Biomedical Engineering, University of West Attica, 12243Athens, Greece
| | - Sara Monticelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Christos Delidakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Evanthia Zacharioudaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| |
Collapse
|
12
|
Troost T, Binshtok U, Sprinzak D, Klein T. Cis-inhibition suppresses basal Notch signaling during sensory organ precursor selection. Proc Natl Acad Sci U S A 2023; 120:e2214535120. [PMID: 37252950 PMCID: PMC10266033 DOI: 10.1073/pnas.2214535120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/29/2023] [Indexed: 06/01/2023] Open
Abstract
The emergence of the sensory organ precursor (SOP) from an equivalence group in Drosophila is a paradigm for studying single-cell fate specification through Notch-mediated lateral inhibition. Yet, it remains unclear how only a single SOP is selected from a relatively large group of cells. We show here that a critical aspect of SOP selection is controlled by cis-inhibition (CI), whereby the Notch ligands, Delta (Dl), cis-inhibit Notch receptors in the same cell. Based on the observation that the mammalian ligand Dl-like 1 cannot cis-inhibit Notch in Drosophila, we probe the role of CI in vivo. We develop a mathematical model for SOP selection where Dl activity is independently regulated by the ubiquitin ligases Neuralized and Mindbomb1. We show theoretically and experimentally that Mindbomb1 induces basal Notch activity, which is suppressed by CI. Our results highlight the trade-off between basal Notch activity and CI as a mechanism for singling out a SOP from a large equivalence group.
Collapse
Affiliation(s)
- Tobias Troost
- Institut fuer Genetik, Heinrich-Heine-Universtitaet Duesseldorf40225Duesseldorf, Germany
| | - Udi Binshtok
- School of Neurobiology, Biochemistry, and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - David Sprinzak
- School of Neurobiology, Biochemistry, and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv69978, Israel
| | - Thomas Klein
- Institut fuer Genetik, Heinrich-Heine-Universtitaet Duesseldorf40225Duesseldorf, Germany
| |
Collapse
|
13
|
The role of Hedgehog and Notch signaling pathway in cancer. MOLECULAR BIOMEDICINE 2022; 3:44. [PMID: 36517618 PMCID: PMC9751255 DOI: 10.1186/s43556-022-00099-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022] Open
Abstract
Notch and Hedgehog signaling are involved in cancer biology and pathology, including the maintenance of tumor cell proliferation, cancer stem-like cells, and the tumor microenvironment. Given the complexity of Notch signaling in tumors, its role as both a tumor promoter and suppressor, and the crosstalk between pathways, the goal of developing clinically safe, effective, tumor-specific Notch-targeted drugs has remained intractable. Drugs developed against the Hedgehog signaling pathway have affirmed definitive therapeutic effects in basal cell carcinoma; however, in some contexts, the challenges of tumor resistance and recurrence leap to the forefront. The efficacy is very limited for other tumor types. In recent years, we have witnessed an exponential increase in the investigation and recognition of the critical roles of the Notch and Hedgehog signaling pathways in cancers, and the crosstalk between these pathways has vast space and value to explore. A series of clinical trials targeting signaling have been launched continually. In this review, we introduce current advances in the understanding of Notch and Hedgehog signaling and the crosstalk between pathways in specific tumor cell populations and microenvironments. Moreover, we also discuss the potential of targeting Notch and Hedgehog for cancer therapy, intending to promote the leap from bench to bedside.
Collapse
|
14
|
Khamaisi B, Luca VC, Blacklow SC, Sprinzak D. Functional Comparison between Endogenous and Synthetic Notch Systems. ACS Synth Biol 2022; 11:3343-3353. [PMID: 36107643 PMCID: PMC9594772 DOI: 10.1021/acssynbio.2c00247] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The Notch pathway converts receptor-ligand interactions at the cell surface into a transcriptional response in the receiver cell. In recent years, synthetic Notch systems (synNotch) that respond to different inputs and transduce different transcriptional responses have been engineered. One class of synNotch systems uses antibody-antigen interactions at the cell surface to induce the proteolytic cleavage cascade of the endogenous Notch autoregulatory core and the consequent release of a synNotch intracellular domain (ICD), converting surface antigen detection into a cellular response. While the activation of endogenous Notch requires ubiquitylation and subsequent endocytosis of the ligand ICD, these synNotch systems do not seem to have such a requirement because the synNotch ligands completely lack an ICD. This observation raises questions about existing models for the synNotch activation mechanism. Here, we test how different structural and biochemical factors affect the dependence of endogenous and synthetic Notch activation on ligand ICD. We compare the behavior of antibody-antigen synNotch (aa-synNotch) to that of endogenous Notch, and to a synNotch system that uses rapamycin induced dimerization of FK506 binding protein (FKBP) and FKBP rapamycin binding (FRB) domaindimerization domains (ff-synNotch), which still requires a ligand ICD. We found that differences in receptor-ligand affinity, in the identity of the transmembrane domain, or in the presence or absence of extracellular epidermal growth factor repeats cannot explain the differences in ligand ICD requirement that distinguishes aa-synNotch from endogenous Notch or ff-synNotch. We also found that unlike endogenous Notch and ff-synNotch, the aa-synNotch system does not exhibit trans-endocytosis of the receptor extracellular domain into the sender cell. These findings suggest that the aa-synNotch systems bypass the ligand ICD requirement because antigen-antibody pairs are able to promote other adhesive cell-cell interactions that provide the mechanical tension needed for ligand activation.
Collapse
Affiliation(s)
- Bassma Khamaisi
- George
S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry,
and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel
| | - Vincent C. Luca
- Department
of Drug Discovery, Moffitt Cancer Center
and Research Institute, Tampa, Florida 33612, United States
| | - Stephen C. Blacklow
- Department
of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - David Sprinzak
- George
S. Wise Faculty of Life Sciences, School of Neurobiology, Biochemistry,
and Biophysics, Tel Aviv University, Tel Aviv 69978, Israel,
| |
Collapse
|
15
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 435] [Impact Index Per Article: 145.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
16
|
Deliconstantinos G, Kalodimou K, Delidakis C. Translational Control of Serrate Expression in Drosophila Cells. In Vivo 2021; 35:859-869. [PMID: 33622878 DOI: 10.21873/invivo.12326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The DSL proteins, Serrate and Delta, which act as Notch receptor ligands, mediate signalling between adjacent cells, when a ligand-expressing cell binds to Notch on an adjacent receiving cell. Notch is ubiquitously expressed and DSL protein mis-expression can have devastating developmental consequences. Although transcriptional regulation of Delta and Serrate has been amply documented, we examined whether they are also regulated at the level of translation. MATERIALS AND METHODS We generated a series of deletions to investigate the initiation codon usage for Serrate using Drosophila S2 cells. RESULTS Serrate mRNA contains three putative ATG initiation codons spanning a 60-codon region upstream of its signal peptide; we found that each one can act as an initiation codon, however, with a different translational efficiency. CONCLUSION Serrate expression is strictly regulated at the translational level.
Collapse
Affiliation(s)
| | - Konstantina Kalodimou
- Institute of Molecular Biology and Biotechnology, F.O.R.T.H., Heraklion, Greece.,Department of Biology, University of Crete Heraklion, Heraklion, Greece
| | - Christos Delidakis
- Institute of Molecular Biology and Biotechnology, F.O.R.T.H., Heraklion, Greece.,Department of Biology, University of Crete Heraklion, Heraklion, Greece
| |
Collapse
|
17
|
Seib E, Klein T. The role of ligand endocytosis in notch signalling. Biol Cell 2021; 113:401-418. [PMID: 34038572 DOI: 10.1111/boc.202100009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
The Notch signalling receptor is a mechanoreceptor that is activated by force. This force elicits a conformational change in Notch that results in the release of its intracellular domain into the cytosol by two consecutive proteolytic cleavages. In most cases, the force is generated by pulling of the ligands on the receptor upon their endocytosis. In this review, we summarise recent work that shed a more detailed light on the role of endocytosis during ligand-dependent Notch activation and discuss the role of ubiquitylation of the ligands during this process.
Collapse
Affiliation(s)
- Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, Duesseldorf, 40225, Germany
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Universitaetsstr. 1, Duesseldorf, 40225, Germany
| |
Collapse
|
18
|
Magadi SS, Voutyraki C, Anagnostopoulos G, Zacharioudaki E, Poutakidou IK, Efraimoglou C, Stapountzi M, Theodorou V, Nikolaou C, Koumbanakis KA, Fullard JF, Delidakis C. Dissecting Hes-centred transcriptional networks in neural stem cell maintenance and tumorigenesis in Drosophila. Development 2020; 147:147/22/dev191544. [PMID: 33229432 DOI: 10.1242/dev.191544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/05/2020] [Indexed: 01/26/2023]
Abstract
Neural stem cells divide during embryogenesis and juvenile life to generate the entire complement of neurons and glia in the nervous system of vertebrates and invertebrates. Studies of the mechanisms controlling the fine balance between neural stem cells and more differentiated progenitors have shown that, in every asymmetric cell division, progenitors send a Delta-Notch signal to their sibling stem cells. Here, we show that excessive activation of Notch or overexpression of its direct targets of the Hes family causes stem-cell hyperplasias in the Drosophila larval central nervous system, which can progress to malignant tumours after allografting to adult hosts. We combined transcriptomic data from these hyperplasias with chromatin occupancy data for Dpn, a Hes transcription factor, to identify genes regulated by Hes factors in this process. We show that the Notch/Hes axis represses a cohort of transcription factor genes. These are excluded from the stem cells and promote early differentiation steps, most likely by preventing the reversion of immature progenitors to a stem-cell fate. We describe the impact of two of these 'anti-stemness' factors, Zfh1 and Gcm, on Notch/Hes-triggered tumorigenesis.
Collapse
Affiliation(s)
- Srivathsa S Magadi
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Chrysanthi Voutyraki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Gerasimos Anagnostopoulos
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Evanthia Zacharioudaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece
| | - Ioanna K Poutakidou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Christina Efraimoglou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Margarita Stapountzi
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece
| | - Vasiliki Theodorou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece
| | - Christoforos Nikolaou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Konstantinos A Koumbanakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece.,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - John F Fullard
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece
| | - Christos Delidakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013 Heraklion, Crete, Greece .,Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| |
Collapse
|
19
|
Unravelling of Hidden Secrets: The Tumour Suppressor Lethal (2) Giant Discs (Lgd)/CC2D1, Notch Signalling and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1287:31-46. [PMID: 33034024 DOI: 10.1007/978-3-030-55031-8_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The endosomal pathway plays a pivotal role upon signal transduction in the Notch pathway. Recent work on lethal (2) giant discs (lgd) points to an additional critical role in avoiding uncontrolled ligand-independent signalling during trafficking of the Notch receptor through the endosomal pathway to the lysosome for degradation. In this chapter, we will outline the journey of Notch through the endosomal system and present an overview of the current knowledge about Lgd and its mammalian orthologs Lgd1/CC2D1b and Lgd2/CC2D1a. We will then discuss how Notch is activated in the absence of lgd function in Drosophila and ask whether there is evidence that a similar ligand-independent activation of the Notch pathway can also happen in mammals if the orthologs are inactivated.
Collapse
|
20
|
Ligand-Induced Cis-Inhibition of Notch Signaling: The Role of an Extracellular Region of Serrate. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:29-49. [PMID: 32072497 DOI: 10.1007/978-3-030-36422-9_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cellular development can be controlled by communication between adjacent cells mediated by the highly conserved Notch signaling system. A cell expressing the Notch receptor on one cell can be activated in trans by ligands on an adjacent cell leading to alteration of transcription and cellular fate. Ligands also have the ability to inhibit Notch signaling, and this can be accomplished when both receptor and ligands are coexpressed in cis on the same cell. The manner in which cis-inhibition is accomplished is not entirely clear but it is known to involve several different protein domains of the ligands and the receptor. Some of the protein domains involved in trans-activation are also used for cis-inhibition, but some are used uniquely for each process. In this work, the involvement of various ligand regions and the receptor are discussed in relation to their contributions to Notch signaling.
Collapse
|
21
|
Antfolk D, Antila C, Kemppainen K, Landor SKJ, Sahlgren C. Decoding the PTM-switchboard of Notch. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118507. [PMID: 31301363 PMCID: PMC7116576 DOI: 10.1016/j.bbamcr.2019.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/03/2019] [Accepted: 07/06/2019] [Indexed: 01/08/2023]
Abstract
The developmentally indispensable Notch pathway exhibits a high grade of pleiotropism in its biological output. Emerging evidence supports the notion of post-translational modifications (PTMs) as a modus operandi controlling dynamic fine-tuning of Notch activity. Although, the intricacy of Notch post-translational regulation, as well as how these modifications lead to multiples of divergent Notch phenotypes is still largely unknown, numerous studies show a correlation between the site of modification and the output. These include glycosylation of the extracellular domain of Notch modulating ligand binding, and phosphorylation of the PEST domain controlling half-life of the intracellular domain of Notch. Furthermore, several reports show that multiple PTMs can act in concert, or compete for the same sites to drive opposite outputs. However, further investigation of the complex PTM crosstalk is required for a complete understanding of the PTM-mediated Notch switchboard. In this review, we aim to provide a consistent and up-to-date summary of the currently known PTMs acting on the Notch signaling pathway, their functions in different contexts, as well as explore their implications in physiology and disease. Furthermore, we give an overview of the present state of PTM research methodology, and allude to a future with PTM-targeted Notch therapeutics.
Collapse
Affiliation(s)
- Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Christian Antila
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Kati Kemppainen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Sebastian K-J Landor
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland; Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
22
|
Wang Z, Kawaguchi K, Honda M, Hashimoto S, Shirasaki T, Okada H, Orita N, Shimakami T, Yamashita T, Sakai Y, Mizukoshi E, Murakami S, Kaneko S. Notch signaling facilitates hepatitis B virus covalently closed circular DNA transcription via cAMP response element-binding protein with E3 ubiquitin ligase-modulation. Sci Rep 2019; 9:1621. [PMID: 30733490 PMCID: PMC6367350 DOI: 10.1038/s41598-018-38139-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Notch1 is regulated by E3 ubiquitin ligases, with proteasomal degradation of the Notch intracellular domain affecting the transcription of target genes. cAMP response element-binding protein (CREB) mediates the transcription of hepatitis B virus (HBV) covalently closed circular DNA (cccDNA). We assessed the relationship between HBV cccDNA and Notch signaling activities. HBV cccDNA levels and relative gene expression were evaluated in HBV-replicating cells treated with Jagged1 shRNA and a γ-secretase inhibitor. The effects of these factors in surgically resected clinical samples were also assessed. Notch inhibition suppressed HBV cccDNA and CREB-related expression but increased ITCH and NUMB levels. Proteasome inhibitor augmented HBV cccDNA, restored Notch and CREB expression, and inhibited ITCH and NUMB function. Increased HBV cccDNA was observed after ITCH and NUMB blockage, even after treatment with the adenylate cyclase activator forskolin; protein kinase A (PKA) inhibitor had the opposite effect. Notch activation and E3 ligase inactivation were observed in HBV-positive cells in clinical liver tissue. Collectively, these findings reveal that Notch signaling activity facilitates HBV cccDNA transcription via CREB to trigger the downstream PKA-phospho-CREB cascade and is regulated by E3 ubiquitin ligase-modulation of the Notch intracellular domain.
Collapse
Affiliation(s)
- Zijing Wang
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Kazunori Kawaguchi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan.
| | - Masao Honda
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Shinichi Hashimoto
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Takayoshi Shirasaki
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hikari Okada
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Noriaki Orita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Tetsuro Shimakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Yoshio Sakai
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Seishi Murakami
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
23
|
Tong K, Wagle M, Guo S. Antibody Uptake Assay in the Embryonic Zebrafish Forebrain to Study Notch Signaling Dynamics in Neural Progenitor Cells In Vivo. Methods Mol Biol 2019; 1576:273-281. [PMID: 28434166 PMCID: PMC5653460 DOI: 10.1007/7651_2017_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cells can generate cell fate heterogeneity through asymmetric cell division (ACD). ACD derives from the asymmetric segregation of fate-determining molecules and/or organelles in the dividing cell. Radial glia in the embryonic zebrafish forebrain are an excellent model for studying the molecular mechanisms regulating ACD of stem cells in vertebrates, especially for live imaging concerning in vivo molecular and cellular dynamics. Due to the current difficulty in expressing fluorescent reporter-tagged proteins at physiological levels in zebrafish for live imaging, we have developed an antibody uptake assay to label proteins in live embryonic zebrafish forebrain with high specificity. DeltaD is a transmembrane ligand in Notch signaling pathway in the context of ACD of radial glia in zebrafish. By using this assay, we have successfully observed the in vivo dynamics of DeltaD for studying ACD of radial glia in the embryonic zebrafish forebrain.
Collapse
Affiliation(s)
- Kai Tong
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai, China
- Department of Bioengineering and Therapeutic Sciences, Programs in Human Genetics and Biological Sciences, ELi and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Mahendra Wagle
- Department of Bioengineering and Therapeutic Sciences, Programs in Human Genetics and Biological Sciences, ELi and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Su Guo
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai, China.
- Department of Bioengineering and Therapeutic Sciences, Programs in Human Genetics and Biological Sciences, ELi and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA.
| |
Collapse
|
24
|
Bigas A, Espinosa L. The multiple usages of Notch signaling in development, cell differentiation and cancer. Curr Opin Cell Biol 2018; 55:1-7. [PMID: 30006050 DOI: 10.1016/j.ceb.2018.06.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
Abstract
Notch is a well-conserved signaling pathway all through evolution that is crucial to specify different cell fates. Although there is a strong context dependent component in each decision, the basic mechanisms that originate from the interplay among ligands and receptors is greatly preserved. In this review we will cover the latest findings on the different mechanisms for Notch activation and signaling. The regulation of this pathway is essential to understand development, cell differentiation and disease.
Collapse
Affiliation(s)
- Anna Bigas
- Program in Cancer Research, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain.
| | - Lluis Espinosa
- Program in Cancer Research, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain.
| |
Collapse
|
25
|
Berndt N, Seib E, Kim S, Troost T, Lyga M, Langenbach J, Haensch S, Kalodimou K, Delidakis C, Klein T. Ubiquitylation-independent activation of Notch signalling by Delta. eLife 2017; 6:27346. [PMID: 28960177 PMCID: PMC5675594 DOI: 10.7554/elife.27346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/28/2017] [Indexed: 12/29/2022] Open
Abstract
Ubiquitylation (ubi) by the E3-ligases Mindbomb1 (Mib1) and Neuralized (Neur) is required for activation of the DSL ligands Delta (Dl) and Serrate (Ser) to activate Notch signalling. These ligases transfer ubiquitin to lysines of the ligands' intracellular domains (ICDs), which sends them into an Epsin-dependent endocytic pathway. Here, we have tested the requirement of ubi of Dl for signalling. We found that Dl requires ubi for its full function, but can also signal in two ubi-independent modes, one dependent and one independent of Neur. We identified two neural lateral specification processes where Dl signals in an ubi-independent manner. Neur, which is needed for these processes, was shown to be able to activate Dl in an ubi-independent manner. Our analysis suggests that one important role of DSL protein ubi by Mib1 is their release from cis-inhibitory interactions with Notch, enabling them to trans-activate Notch on adjacent cells.
Collapse
Affiliation(s)
- Nicole Berndt
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Ekaterina Seib
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Soya Kim
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany.,Molekulare Zellbiologie, Institut I für Anatomie, Uniklinik Köln, Universität zu Köln, Köln, Germany
| | - Tobias Troost
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Marvin Lyga
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Jessica Langenbach
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Sebastian Haensch
- Center of Advanced Imaging, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| | - Konstantina Kalodimou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.,Department of Biology, University of Crete, Heraklion, Greece
| | - Christos Delidakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.,Department of Biology, University of Crete, Heraklion, Greece
| | - Thomas Klein
- Institute of Genetics, Heinrich-Heine-Universitaet Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
26
|
Perez-Mockus G, Schweisguth F. Cell Polarity and Notch Signaling: Linked by the E3 Ubiquitin Ligase Neuralized? Bioessays 2017; 39. [DOI: 10.1002/bies.201700128] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/17/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Gantas Perez-Mockus
- Institut Pasteur,; Dept of Developmental and Stem Cell Biology; F-75015 Paris France
- CNRS; UMR3738; F-75015 Paris France
- Univ. Pierre et Marie Curie; Cellule Pasteur UPMC; rue du Dr Roux 75015 Paris France
| | - Francois Schweisguth
- Institut Pasteur,; Dept of Developmental and Stem Cell Biology; F-75015 Paris France
- CNRS; UMR3738; F-75015 Paris France
| |
Collapse
|
27
|
Perez-Mockus G, Roca V, Mazouni K, Schweisguth F. Neuralized regulates Crumbs endocytosis and epithelium morphogenesis via specific Stardust isoforms. J Cell Biol 2017; 216:1405-1420. [PMID: 28400441 PMCID: PMC5412571 DOI: 10.1083/jcb.201611196] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 11/22/2022] Open
Abstract
The E3 ubiquitin ligase Neuralized is shown to interact with a subset of the Stardust isoforms to regulate the endocytosis of the apical protein Crumbs and thereby promote epithelial remodeling during Drosophila development. Crumbs (Crb) is a conserved determinant of apical membrane identity that regulates epithelial morphogenesis in many developmental contexts. In this study, we identify the Crb complex protein Stardust (Sdt) as a target of the E3 ubiquitin ligase Neuralized (Neur) in Drosophila melanogaster. Neur interacts with and down-regulates specific Sdt isoforms containing a Neur binding motif (NBM). Using a CRISPR (clustered regularly interspaced short palindromic repeats)-induced deletion of the NBM-encoding exon, we found that Sdt is a key Neur target and that Neur acts via Sdt to down-regulate Crb. We further show that Neur promotes the endocytosis of Crb via the NBM-containing isoforms of Sdt. Although the regulation of Crb by Neur is not strictly essential, it contributes to epithelium remodeling in the posterior midgut and thereby facilitates the trans-epithelial migration of the primordial germ cells in early embryos. Thus, our study uncovers a novel regulatory mechanism for the developmental control of Crb-mediated morphogenesis.
Collapse
Affiliation(s)
- Gantas Perez-Mockus
- Department of Developmental and Stem Cell Biology, Institut Pasteur, F-75015 Paris, France.,Centre National de la Recherché Scientifique, UMR3738, F-75015 Paris, France.,Cellule Pasteur, Université Pierre et Marie Curie, F-75015 Paris, France
| | - Vanessa Roca
- Department of Developmental and Stem Cell Biology, Institut Pasteur, F-75015 Paris, France.,Centre National de la Recherché Scientifique, UMR3738, F-75015 Paris, France
| | - Khalil Mazouni
- Department of Developmental and Stem Cell Biology, Institut Pasteur, F-75015 Paris, France.,Centre National de la Recherché Scientifique, UMR3738, F-75015 Paris, France
| | - François Schweisguth
- Department of Developmental and Stem Cell Biology, Institut Pasteur, F-75015 Paris, France .,Centre National de la Recherché Scientifique, UMR3738, F-75015 Paris, France
| |
Collapse
|
28
|
Domsch K, Acs A, Obermeier C, Nguyen HT, Reim I. Identification of the essential protein domains for Mib2 function during the development of the Drosophila larval musculature and adult flight muscles. PLoS One 2017; 12:e0173733. [PMID: 28282454 PMCID: PMC5345843 DOI: 10.1371/journal.pone.0173733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/24/2017] [Indexed: 12/01/2022] Open
Abstract
The proper differentiation and maintenance of myofibers is fundamental to a functional musculature. Disruption of numerous mostly structural factors leads to perturbations of these processes. Among the limited number of known regulatory factors for these processes is Mind bomb2 (Mib2), a muscle-associated E3 ubiquitin ligase, which was previously established to be required for maintaining the integrity of larval muscles. In this study, we have examined the mechanistic aspects of Mib2 function by performing a detailed functional dissection of the Mib2 protein. We show that the ankyrin repeats, in its entirety, and the hitherto uncharacterized Mib-specific domains (MIB), are important for the major function of Mib2 in skeletal and visceral muscles in the Drosophila embryo. Furthermore, we characterize novel mib2 alleles that have arisen from a forward genetic screen aimed at identifying regulators of myogenesis. Two of these alleles are viable, but flightless hypomorphic mib2 mutants, and harbor missense mutations in the MIB domain and RING finger, respectively. Functional analysis of these new alleles, including in vivo imaging, demonstrates that Mib2 plays an additional important role in the development of adult thorax muscles, particularly in maintaining the larval templates for the dorsal longitudinal indirect flight muscles during metamorphosis.
Collapse
Affiliation(s)
- Katrin Domsch
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Acs
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Claudia Obermeier
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hanh T. Nguyen
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ingolf Reim
- Department of Biology, Division of Developmental Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
29
|
Abstract
The highly conserved Notch signalling pathway functions in many different developmental and homeostatic processes, which raises the question of how this pathway can achieve such diverse outcomes. With a direct route from the membrane to the nucleus, the Notch pathway has fewer opportunities for regulation than do many other signalling pathways, yet it generates exquisitely patterned structures, including sensory hair cells and branched arterial networks. More confusingly, its activity promotes tissue growth and cancers in some circumstances but cell death and tumour suppression in others. Many different regulatory mechanisms help to shape the activity of the Notch pathway, generating functional outputs that are appropriate for each context. These mechanisms include the receptor-ligand landscape, the tissue topology, the nuclear environment and the connectivity of the regulatory networks.
Collapse
Affiliation(s)
- Sarah J Bray
- Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
30
|
Liu J, Shen JX, Wen XF, Guo YX, Zhang GJ. Targeting Notch degradation system provides promise for breast cancer therapeutics. Crit Rev Oncol Hematol 2016; 104:21-9. [PMID: 27263934 DOI: 10.1016/j.critrevonc.2016.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 04/18/2016] [Accepted: 05/19/2016] [Indexed: 02/05/2023] Open
Abstract
Notch receptor signaling pathways play an important role, not only in normal breast development but also in breast cancer development and progression. As a group of ligand-induced proteins, different subtypes of mammalian Notch (Notch1-4) are sensitive to subtle changes in protein levels. Thus, a clear understanding of mechanisms of Notch protein turnover is essential for understanding normal and pathological mechanisms of Notch functions. It has been suggested that there is a close relationship between the carcinogenesis and the dysregulation of Notch degradation. However, this relationship remains mostly undefined in the context of breast cancer, as protein degradation is mediated by numerous signaling pathways as well as certain molecule modulators (activators/inhibitors). In this review, we summarize the published data regarding the regulation of Notch family member degradation in breast cancer, while emphasizing areas that are likely to provide new therapeutic modalities for mechanism-based anti-cancer drugs.
Collapse
Affiliation(s)
- Jing Liu
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Jia-Xin Shen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Xiao-Fen Wen
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Yu-Xian Guo
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| | - Guo-Jun Zhang
- Chang Jiang Scholar's Laboratory, Shantou University Medical College, Shantou, Guangdong Province, PR China; Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China; The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong Province, PR China.
| |
Collapse
|
31
|
Structure and function of the Mind bomb E3 ligase in the context of Notch signal transduction. Curr Opin Struct Biol 2016; 41:38-45. [PMID: 27285058 DOI: 10.1016/j.sbi.2016.05.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/16/2022]
Abstract
The Notch signaling pathway has a critical role in cell fate determination and tissue homeostasis in a variety of different lineages. In the context of normal Notch signaling, the Notch receptor of the 'signal-receiving' cell is activated in trans by a Notch ligand from a neighboring 'signal-sending' cell. Genetic studies in several model organisms have established that ubiquitination of the Notch ligand, and its regulated endocytosis, is essential for transmission of this activation signal. In mammals, this ubiquitination step is dependent on the protein Mind bomb 1 (Mib1), a large multi-domain RING-type E3 ligase, and its direct interaction with the intracellular tails of Notch ligand molecules. Here, we discuss our current understanding of Mind bomb structure and mechanism in the context of Notch signaling and beyond.
Collapse
|
32
|
Okano M, Matsuo H, Nishimura Y, Hozumi K, Yoshioka S, Tonoki A, Itoh M. Mib1 modulates dynamin 2 recruitment via Snx18 to promote Dll1 endocytosis for efficient Notch signaling. Genes Cells 2016; 21:425-41. [DOI: 10.1111/gtc.12350] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 01/28/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Makoto Okano
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Hiromi Matsuo
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Yuya Nishimura
- Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Katsuto Hozumi
- Department of Immunology; Tokai University School of Medicine; Isehara Kanagawa 259-1193 Japan
| | - Saho Yoshioka
- Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Ayako Tonoki
- Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| | - Motoyuki Itoh
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
- Graduate School of Pharmaceutical Sciences; Chiba University; Chiba 260-8675 Japan
| |
Collapse
|
33
|
Imai Y, Kobayashi Y, Inoshita T, Meng H, Arano T, Uemura K, Asano T, Yoshimi K, Zhang CL, Matsumoto G, Ohtsuka T, Kageyama R, Kiyonari H, Shioi G, Nukina N, Hattori N, Takahashi R. The Parkinson's Disease-Associated Protein Kinase LRRK2 Modulates Notch Signaling through the Endosomal Pathway. PLoS Genet 2015; 11:e1005503. [PMID: 26355680 PMCID: PMC4565672 DOI: 10.1371/journal.pgen.1005503] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/14/2015] [Indexed: 12/03/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of familial and idiopathic Parkinson’s disease (PD). We have identified two novel LRRK2-associated proteins, a HECT-type ubiquitin ligase, HERC2, and an adaptor-like protein with six repeated Neuralized domains, NEURL4. LRRK2 binds to NEURL4 and HERC2 via the LRRK2 Ras of complex proteins (ROC) domain and NEURL4, respectively. HERC2 and NEURL4 link LRRK2 to the cellular vesicle transport pathway and Notch signaling, through which the LRRK2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. This process negatively regulates Notch signaling through cis-inhibition by stabilizing Dll1/Dl, which accelerates neural stem cell differentiation and modulates the function and survival of differentiated dopaminergic neurons. These effects are strengthened by the R1441G ROC domain-mutant of LRRK2. These findings suggest that the alteration of Notch signaling in mature neurons is a component of PD etiology linked to LRRK2. LRRK2 is linked to autosomal dominant late-onset Parkinson’s disease, suggesting that LRRK2 gain-of-function mutations lead to age-dependent degeneration of the midbrain dopaminergic neurons. In this study, we describe two novel LRRK2-associated proteins HERC2 and NEURL4, which are a ubiquitin ligase and an adaptor-like protein, respectively. HERC2 and NEURL4 direct LRRK2 to Notch signaling pathway, in which the LRRK2-NEURL4-HERC2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. As a result, the amounts of Dll1/D1 on the plasma membrane are increased, which affects negatively Notch signaling through cis-inhibition. The effect is enhanced by a Parkinson’s-disease associated mutation of LRRK2. Inhibition of Notch signaling in adult dopaminergic neurons impairs its functions and survival. These findings indicate a possible link between Notch pathway and Parkinson’s disease.
Collapse
Affiliation(s)
- Yuzuru Imai
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- * E-mail: (YI); (NH); (RT)
| | - Yoshito Kobayashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
| | - Tsuyoshi Inoshita
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hongrui Meng
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Taku Arano
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kengo Uemura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Asano
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kenji Yoshimi
- Department of Neurophysiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chang-Liang Zhang
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
| | - Gen Matsumoto
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiyuki Ohtsuka
- Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Ryoichiro Kageyama
- Department of Cell Biology, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Go Shioi
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Japan
| | - Nobuyuki Nukina
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- * E-mail: (YI); (NH); (RT)
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- CREST (Core Research for Evolutionary Science and Technology), Japan Science and Technology Agency, Saitama, Japan
- * E-mail: (YI); (NH); (RT)
| |
Collapse
|
34
|
Pandya K, Wyatt D, Gallagher B, Shah D, Baker A, Bloodworth J, Zlobin A, Pannuti A, Green A, Ellis IO, Filipovic A, Sagert J, Rana A, Albain KS, Miele L, Denning MF, Osipo C. PKCα Attenuates Jagged-1-Mediated Notch Signaling in ErbB-2-Positive Breast Cancer to Reverse Trastuzumab Resistance. Clin Cancer Res 2015; 22:175-86. [PMID: 26350262 DOI: 10.1158/1078-0432.ccr-15-0179] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023]
Abstract
PURPOSE Breast cancer is the second leading cause of cancer mortality among women worldwide. The major problem with current treatments is tumor resistance, recurrence, and disease progression. ErbB-2-positive breast tumors are aggressive and frequently become resistant to trastuzumab or lapatinib. We showed previously that Notch-1 is required for trastuzumab resistance in ErbB-2-positive breast cancer. EXPERIMENTAL DESIGN Here, we sought to elucidate mechanisms by which ErbB-2 attenuates Notch signaling and how this is reversed by trastuzumab or lapatinib. RESULTS The current study elucidates a novel Notch inhibitory mechanism by which PKCα downstream of ErbB-2 (i) restricts the availability of Jagged-1 at the cell surface to transactivate Notch, (ii) restricts the critical interaction between Jagged-1 and Mindbomb-1, an E3 ligase that is required for Jagged-1 ubiquitinylation and subsequent Notch activation, (iii) reverses trastuzumab resistance in vivo, and (iv) predicts better outcome in women with ErbB-2-positive breast cancer. CONCLUSIONS The clinical impact of these studies is PKCα is potentially a good prognostic marker for low Notch activity and increased trastuzumab sensitivity in ErbB-2-positive breast cancer. Moreover, women with ErbB-2-positive breast tumors expressing high Notch activation and low PKCα expression could be the best candidates for anti-Notch therapy.
Collapse
Affiliation(s)
- Kinnari Pandya
- Molecular Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Debra Wyatt
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | - Brian Gallagher
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | - Deep Shah
- Department of Molecular Pharmacology and Therapeutics, Louisiana State University, New Orleans, Louisiana
| | - Andrew Baker
- Integrated Cell Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Jeffrey Bloodworth
- Molecular Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Andrei Zlobin
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | | | - Andrew Green
- Department of Histopathology, University of Nottingham and University Hospital NHS Trust, Nottingham, United Kingdom
| | - Ian O Ellis
- Department of Histopathology, University of Nottingham and University Hospital NHS Trust, Nottingham, United Kingdom
| | | | | | - Ajay Rana
- Department of Molecular Pharmacology and Therapeutics, Louisiana State University, New Orleans, Louisiana
| | - Kathy S Albain
- Department of Medicine/Hematology and Oncology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois
| | - Lucio Miele
- Louisiana State University, New Orleans, Louisiana
| | - Mitchell F Denning
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana. Department of Pathology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois
| | - Clodia Osipo
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana. Department of Pathology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois. Department of Microbiology and Immunology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois.
| |
Collapse
|
35
|
McMillan BJ, Schnute B, Ohlenhard N, Zimmerman B, Miles L, Beglova N, Klein T, Blacklow SC. A tail of two sites: a bipartite mechanism for recognition of notch ligands by mind bomb E3 ligases. Mol Cell 2015; 57:912-924. [PMID: 25747658 DOI: 10.1016/j.molcel.2015.01.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 12/22/2014] [Accepted: 01/09/2015] [Indexed: 12/30/2022]
Abstract
Mind bomb (Mib) proteins are large, multi-domain E3 ligases that promote ubiquitination of the cytoplasmic tails of Notch ligands. This ubiquitination step marks the ligand proteins for epsin-dependent endocytosis, which is critical for in vivo Notch receptor activation. We present here crystal structures of the substrate recognition domains of Mib1, both in isolation and in complex with peptides derived from Notch ligands. The structures, in combination with biochemical, cellular, and in vivo assays, show that Mib1 contains two independent substrate recognition domains that engage two distinct epitopes from the cytoplasmic tail of the ligand Jagged1, one in the intracellular membrane proximal region and the other near the C terminus. Together, these studies provide insights into the mechanism of ubiquitin transfer by Mind bomb E3 ligases, illuminate a key event in ligand-induced activation of Notch receptors, and identify a potential target for therapeutic modulation of Notch signal transduction in disease.
Collapse
Affiliation(s)
| | | | | | - Brandon Zimmerman
- Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Boston, MA 02215 USA
| | - Laura Miles
- Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Boston, MA 02215 USA
| | - Natalia Beglova
- Harvard Medical School, Boston, MA 02115, USA; Beth-Israel Deaconess Medical Center, Boston, MA, 02215 USA
| | - Thomas Klein
- University of Dusseldorf, Dusseldorf 40225, Germany
| | - Stephen C Blacklow
- Harvard Medical School, Boston, MA 02115, USA; Dana Farber Cancer Institute, Boston, MA 02215 USA.
| |
Collapse
|
36
|
Identification of the Mind Bomb1 Interaction Domain in Zebrafish DeltaD. PLoS One 2015; 10:e0127864. [PMID: 26020642 PMCID: PMC4447371 DOI: 10.1371/journal.pone.0127864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/09/2015] [Indexed: 11/19/2022] Open
Abstract
Ubiquitylation promotes endocytosis of the Notch ligands like Delta and Serrate and is essential for them to effectively activate Notch in a neighboring cell. The RING E3 ligase Mind bomb1 (Mib1) ubiquitylates DeltaD to facilitate Notch signaling in zebrafish. We have identified a domain in the intracellular part of the zebrafish Notch ligand DeltaD that is essential for effective interactions with Mib1. We show that elimination of the Mind bomb1 Interaction Domain (MID) or mutation of specific conserved motifs in this domain prevents effective Mib1-mediated ubiquitylation and internalization of DeltaD. Lateral inhibition mediated by Notch signaling regulates early neurogenesis in zebrafish. In this context, Notch activation suppresses neurogenesis, while loss of Notch-mediated lateral inhibition results in a neurogenic phenotype, where too many cells are allowed to become neurons. While Mib1-mediated endocytosis of DeltaD is essential for effective activation of Notch in a neighboring cell (in trans) it is not required for DeltaD to inhibit function of Notch receptors in the same cell (in cis). As a result, forms of DeltaD that have the MID can activate Notch in trans and suppress early neurogenesis when mRNA encoding it is ectopically expressed in zebrafish embryos. On the other hand, when the MID is eliminated/mutated in DeltaD, its ability to activate Notch in trans fails but ability to inhibit in cis is retained. As a result, ectopic expression of DeltaD lacking an effective MID results in a failure of Notch-mediated lateral inhibition and a neurogenic phenotype.
Collapse
|
37
|
Mertz J, Tan H, Pagala V, Bai B, Chen PC, Li Y, Cho JH, Shaw T, Wang X, Peng J. Sequential Elution Interactome Analysis of the Mind Bomb 1 Ubiquitin Ligase Reveals a Novel Role in Dendritic Spine Outgrowth. Mol Cell Proteomics 2015; 14:1898-910. [PMID: 25931508 DOI: 10.1074/mcp.m114.045898] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Indexed: 11/06/2022] Open
Abstract
The mind bomb 1 (Mib1) ubiquitin ligase is essential for controlling metazoan development by Notch signaling and possibly the Wnt pathway. It is also expressed in postmitotic neurons and regulates neuronal morphogenesis and synaptic activity by mechanisms that are largely unknown. We sought to comprehensively characterize the Mib1 interactome and study its potential function in neuron development utilizing a novel sequential elution strategy for affinity purification, in which Mib1 binding proteins were eluted under different stringency and then quantified by the isobaric labeling method. The strategy identified the Mib1 interactome with both deep coverage and the ability to distinguish high-affinity partners from low-affinity partners. A total of 817 proteins were identified during the Mib1 affinity purification, including 56 high-affinity partners and 335 low-affinity partners, whereas the remaining 426 proteins are likely copurified contaminants or extremely weak binding proteins. The analysis detected all previously known Mib1-interacting proteins and revealed a large number of novel components involved in Notch and Wnt pathways, endocytosis and vesicle transport, the ubiquitin-proteasome system, cellular morphogenesis, and synaptic activities. Immunofluorescence studies further showed colocalization of Mib1 with five selected proteins: the Usp9x (FAM) deubiquitinating enzyme, alpha-, beta-, and delta-catenins, and CDKL5. Mutations of CDKL5 are associated with early infantile epileptic encephalopathy-2 (EIEE2), a severe form of mental retardation. We found that the expression of Mib1 down-regulated the protein level of CDKL5 by ubiquitination, and antagonized CDKL5 function during the formation of dendritic spines. Thus, the sequential elution strategy enables biochemical characterization of protein interactomes; and Mib1 analysis provides a comprehensive interactome for investigating its role in signaling networks and neuronal development.
Collapse
Affiliation(s)
- Joseph Mertz
- From the ‡Departments of Structural Biology and Developmental Neurobiology
| | | | | | - Bing Bai
- From the ‡Departments of Structural Biology and Developmental Neurobiology
| | - Ping-Chung Chen
- From the ‡Departments of Structural Biology and Developmental Neurobiology
| | - Yuxin Li
- From the ‡Departments of Structural Biology and Developmental Neurobiology
| | | | - Timothy Shaw
- §St. Jude Proteomics Facility, ¶Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | | | - Junmin Peng
- From the ‡Departments of Structural Biology and Developmental Neurobiology, §St. Jude Proteomics Facility,
| |
Collapse
|
38
|
Čajánek L, Glatter T, Nigg EA. The E3 ubiquitin ligase Mib1 regulates Plk4 and centriole biogenesis. J Cell Sci 2015; 128:1674-82. [PMID: 25795303 DOI: 10.1242/jcs.166496] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/18/2015] [Indexed: 02/02/2023] Open
Abstract
Centrioles function as core components of centrosomes and as basal bodies for the formation of cilia and flagella. Thus, effective control of centriole numbers is essential for embryogenesis, tissue homeostasis and genome stability. In mammalian cells, the centriole duplication cycle is governed by Polo-like kinase 4 (Plk4). Here, we identify the E3 ubiquitin ligase Mind bomb (Mib1) as a new interaction partner of Plk4. We show that Mib1 localizes to centriolar satellites but redistributes to centrioles in response to conditions that induce centriole amplification. The E3 ligase activity of Mib1 triggers ubiquitylation of Plk4 on multiple sites, causing the formation of Lys11-, Lys29- and Lys48-ubiquitin linkages. These modifications control the abundance of Plk4 and its ability to interact with centrosomal proteins, thus counteracting centriole amplification induced by excess Plk4. Collectively, these results identify the interaction between Mib1 and Plk4 as a new and important element in the control of centriole homeostasis.
Collapse
Affiliation(s)
- Lukas Čajánek
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Timo Glatter
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Erich A Nigg
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| |
Collapse
|
39
|
Kiparaki M, Zarifi I, Delidakis C. bHLH proteins involved in Drosophila neurogenesis are mutually regulated at the level of stability. Nucleic Acids Res 2015; 43:2543-59. [PMID: 25694512 PMCID: PMC4357701 DOI: 10.1093/nar/gkv083] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Proneural bHLH activators are expressed in all neuroectodermal regions prefiguring events of central and peripheral neurogenesis. Drosophila Sc is a prototypical proneural activator that heterodimerizes with the E-protein Daughterless (Da) and is antagonized by, among others, the E(spl) repressors. We determined parameters that regulate Sc stability in Drosophila S2 cells. We found that Sc is a very labile phosphoprotein and its turnover takes place via at least three proteasome-dependent mechanisms. (i) When Sc is in excess of Da, its degradation is promoted via its transactivation domain (TAD). (ii) In a DNA-bound Da/Sc heterodimer, Sc degradation is promoted via an SPTSS phosphorylation motif and the AD1 TAD of Da; Da is spared in the process. (iii) When E(spl)m7 is expressed, it complexes with Sc or Da/Sc and promotes their degradation in a manner that requires the corepressor Groucho and the Sc SPTSS motif. Da/Sc reciprocally promotes E(spl)m7 degradation. Since E(spl)m7 is a direct target of Notch, the mutual destabilization of Sc and E(spl) may contribute in part to the highly conserved anti-neural activity of Notch. Sc variants lacking the SPTSS motif are dramatically stabilized and are hyperactive in transgenic flies. Our results propose a novel mechanism of regulation of neurogenesis, involving the stability of key players in the process.
Collapse
Affiliation(s)
- Marianthi Kiparaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, and Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Ioanna Zarifi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, and Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| | - Christos Delidakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, and Department of Biology, University of Crete, 70013 Heraklion, Crete, Greece
| |
Collapse
|
40
|
Schweisguth F. Asymmetric cell division in the Drosophila bristle lineage: from the polarization of sensory organ precursor cells to Notch-mediated binary fate decision. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:299-309. [PMID: 25619594 PMCID: PMC4671255 DOI: 10.1002/wdev.175] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 12/11/2014] [Accepted: 12/19/2014] [Indexed: 12/26/2022]
Abstract
Asymmetric cell division (ACD) is a simple and evolutionary conserved process whereby a mother divides to generate two daughter cells with distinct developmental potentials. This process can generate cell fate diversity during development. Fate asymmetry may result from the unequal segregation of molecules and/or organelles between the two daughter cells. Here, I will review how fate asymmetry is regulated in the sensory bristle lineage in Drosophila and focus on the molecular mechanisms underlying ACD of the sensory organ precursor cells (SOPs). WIREs Dev Biol 2015, 4:299–309. doi: 10.1002/wdev.175 For further resources related to this article, please visit theWIREs website. Conflict of interest: The author has declared no conflicts of interest for this article.
Collapse
|
41
|
Zazueta-Novoa V, Wessel GM. Protein degradation machinery is present broadly during early development in the sea urchin. Gene Expr Patterns 2014; 15:135-41. [PMID: 24963879 DOI: 10.1016/j.gep.2014.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/03/2014] [Accepted: 06/05/2014] [Indexed: 11/29/2022]
Abstract
Ubiquitin-dependent proteosome-mediated proteolysis is an important pathway of degradation that controls the timed destruction of cellular proteins in all tissues. All intracellular proteins and many extracellular proteins are continually being hydrolyzed to their constituent amino acids as a result of their recognition by E3 ligases for specific targeting of ubiquitination. Gustavus is a member of an ECS-type E3 ligase which interacts with Vasa, a DEAD-box RNA helicase, to regulate its localization during sea urchin embryonic development, and Gustavus mRNA accumulation is highly localized and dynamic during development. We tested if the core complex for Gustavus function was present in the embryo and if other SOCS box proteins also had restricted expression profiles that would inform future research. Expression patterns of the key members of the proteasomal function, such as the E3 core complex which interacts with Gustavus, and other E3-SOCS box proteins, are widely spread and dynamic in early development of the embryo suggesting broad core complex availability in the proteasome degradation pathway and temporal/spatial enrichments of various E3 ligase dependent targeting mechanisms.
Collapse
Affiliation(s)
- Vanesa Zazueta-Novoa
- Department of Molecular and Cellular Biology & Biochemistry, Brown University, Providence, RI 02912, USA
| | - Gary M Wessel
- Department of Molecular and Cellular Biology & Biochemistry, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
42
|
Saritas-Yildirim B, Silva EM. The role of targeted protein degradation in early neural development. Genesis 2014; 52:287-99. [PMID: 24623518 DOI: 10.1002/dvg.22771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 11/08/2022]
Abstract
As neural stem cells differentiate into neurons during neurogenesis, the proteome of the cells is restructured by de novo expression and selective removal of regulatory proteins. The control of neurogenesis at the level of gene regulation is well documented and the regulation of protein abundance through protein degradation via the Ubiquitin/26S proteasome pathway is a rapidly developing field. This review describes our current understanding of the role of the proteasome pathway in neurogenesis. Collectively, the studies show that targeted protein degradation is an important regulatory mechanism in the generation of new neurons.
Collapse
|
43
|
Tognon E, Vaccari T. Immunohistochemical tools and techniques to visualize Notch in Drosophila melanogaster. Methods Mol Biol 2014; 1187:63-78. [PMID: 25053481 DOI: 10.1007/978-1-4939-1139-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The ability to accurately visualize proteins in Drosophila tissues is critical for studying their abundance and localization relative to the morphology of cells during tissue development and homeostasis. Here we describe the procedure to visualize Notch localization in whole-mount preparations of several Drosophila organs using confocal microscopy. The use of monoclonal antibodies directed to distinct portions of Notch allows one to follow the fate of the receptor during constitutive and inductive processes. The protocol described here can be used to co-label with antibodies recognizing markers of subcellular compartments in wild-type as well as mutant tissues.
Collapse
Affiliation(s)
- Emiliana Tognon
- Istituto FIRC di Oncologia Molecolare (IFOM), IFOM-IEO Campus, via Adamello 16, 20139, Milano, Italy
| | | |
Collapse
|
44
|
Couturier L, Schweisguth F. Antibody uptake assay and in vivo imaging to study intracellular trafficking of Notch and Delta in Drosophila. Methods Mol Biol 2014; 1187:79-86. [PMID: 25053482 DOI: 10.1007/978-1-4939-1139-4_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Notch signaling depends on regulated intracellular trafficking of the receptor and its ligands (Kopan and Ilagan, Cell 137:216-233, 2009; Le Borgne et al., Development 132:1751-1762, 2005). Here we describe two methods to study the intracellular trafficking of Notch and Delta in Drosophila. First, an ex vivo antibody uptake assay is used to monitor endocytosis of Notch and Delta by living cells in dissected explants (Le Borgne and Schweisguth, Dev Cell 5:139-148, 2003). Second, real-time imaging of fluorescent proteins that are expressed at physiological levels is used to study trafficking of Notch in living flies (Venken et al., Science 314:1747-1751, 2006; Couturier et al., Nat Cell Biol 14, 131-139, 2012).
Collapse
Affiliation(s)
- Lydie Couturier
- Département de Biologie du Développement, Unité de Génétique du Développement de la Drosophile, Institut Pasteur and CNRS, 25 rue du Dr Roux, 75015, Paris, France
| | | |
Collapse
|
45
|
Ubiquitinations in the notch signaling pathway. Int J Mol Sci 2013; 14:6359-81. [PMID: 23519106 PMCID: PMC3634445 DOI: 10.3390/ijms14036359] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/11/2013] [Accepted: 03/14/2013] [Indexed: 12/22/2022] Open
Abstract
The very conserved Notch pathway is used iteratively during development and adulthood to regulate cell fates. Notch activation relies on interactions between neighboring cells, through the binding of Notch receptors to their ligands, both transmembrane molecules. This inter-cellular contact initiates a cascade of events eventually transforming the cell surface receptor into a nuclear factor acting on the transcription of specific target genes. This review highlights how the various processes undergone by Notch receptors and ligands that regulate the pathway are linked to ubiquitination events.
Collapse
|
46
|
Breaking it down: the ubiquitin proteasome system in neuronal morphogenesis. Neural Plast 2013; 2013:196848. [PMID: 23476809 PMCID: PMC3586504 DOI: 10.1155/2013/196848] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/31/2012] [Indexed: 01/20/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) is most widely known for its role in intracellular protein degradation; however, in the decades since its discovery, ubiquitination has been associated with the regulation of a wide variety of cellular processes. The addition of ubiquitin tags, either as single moieties or as polyubiquitin chains, has been shown not only to mediate degradation by the proteasome and the lysosome, but also to modulate protein function, localization, and endocytosis. The UPS plays a particularly important role in neurons, where local synthesis and degradation work to balance synaptic protein levels at synapses distant from the cell body. In recent years, the UPS has come under increasing scrutiny in neurons, as elements of the UPS have been found to regulate such diverse neuronal functions as synaptic strength, homeostatic plasticity, axon guidance, and neurite outgrowth. Here we focus on recent advances detailing the roles of the UPS in regulating the morphogenesis of axons, dendrites, and dendritic spines, with an emphasis on E3 ubiquitin ligases and their identified regulatory targets.
Collapse
|
47
|
Functional analysis of the NHR2 domain indicates that oligomerization of Neuralized regulates ubiquitination and endocytosis of Delta during Notch signaling. Mol Cell Biol 2012; 32:4933-45. [PMID: 23045391 DOI: 10.1128/mcb.00711-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The Notch pathway plays an integral role in development by regulating cell fate in a wide variety of multicellular organisms. A critical step in the activation of Notch signaling is the endocytosis of the Notch ligands Delta and Serrate. Ligand endocytosis is regulated by one of two E3 ubiquitin ligases, Neuralized (Neur) or Mind bomb. Neur is comprised of a C-terminal RING domain, which is required for Delta ubiquitination, and two Neur homology repeat (NHR) domains. We have previously shown that the NHR1 domain is required for Delta trafficking. Here we show that the NHR1 domain also affects the binding and internalization of Serrate. Furthermore, we show that the NHR2 domain is required for Neur function and that a point mutation in the NHR2 domain (Gly430) abolishes Neur ubiquitination activity and affects ligand internalization. Finally, we provide evidence that Neur can form oligomers in both cultured cells and fly tissues, which regulate Neur activity and, by extension, ligand internalization.
Collapse
|
48
|
Clague M, Liu H, Urbé S. Governance of Endocytic Trafficking and Signaling by Reversible Ubiquitylation. Dev Cell 2012; 23:457-67. [DOI: 10.1016/j.devcel.2012.08.011] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 06/27/2012] [Accepted: 08/21/2012] [Indexed: 12/17/2022]
|
49
|
Kandachar V, Roegiers F. Endocytosis and control of Notch signaling. Curr Opin Cell Biol 2012; 24:534-40. [PMID: 22818956 DOI: 10.1016/j.ceb.2012.06.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/19/2012] [Accepted: 06/21/2012] [Indexed: 02/06/2023]
Abstract
The Notch signaling pathway controls patterning and cell fate decisions during development in metazoans, and is associated with human diseases such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and certain cancers. Studies over the last several years have revealed sophisticated regulation of both the membrane-bound Notch receptor and its ligands by vesicle trafficking. This is perhaps most evident in neural progenitor cells in Drosophila, which divide asymmetrically to segregate Numb, an endocytic adaptor protein that acts as a Notch pathway inhibitor, to one daughter cell. Here, we discuss recent findings addressing how receptor and ligand trafficking to specific membrane compartments control activation of the Notch pathway in asymmetrically dividing cells and other tissues.
Collapse
Affiliation(s)
- Vasundhara Kandachar
- Program in Cancer Biology, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| | | |
Collapse
|
50
|
Shah DK, Zúñiga-Pflücker JC. Notch receptor-ligand interactions during T cell development, a ligand endocytosis-driven mechanism. Curr Top Microbiol Immunol 2012; 360:19-46. [PMID: 22581027 DOI: 10.1007/82_2012_225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling plays an important role during the development of different cell types and tissues. The role of Notch signaling in lymphocyte development, in particular in the development and commitment to the T cell lineage, has been the focus of research for many years. Notch signaling is absolutely required during the commitment and early stages of T cell development. Activation of the Notch signaling pathway is initiated by ligand-receptor interactions and appears to require active endocytosis of Notch ligands. Studies addressing the mechanism underlying endocytosis of Notch ligands have helped to identify the main players important and necessary for this process. Here, we review the Notch ligands, and the proposed models of Notch activation by Notch ligand endocytosis, highlighting key molecules involved. In particular, we discuss recent studies on Notch ligands involved in T cell development, current studies aimed at elucidating the relevance of Notch ligand endocytosis during T cell development and the identification of key players necessary for ligand endocytosis in the thymus and during T cell development.
Collapse
Affiliation(s)
- Divya K Shah
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, M4 N 3M5, Canada.
| | | |
Collapse
|