1
|
Moreira JF, Solá S. Dynamics of Neurogenic Signals as Biological Switchers of Brain Plasticity. Stem Cell Rev Rep 2024:10.1007/s12015-024-10788-2. [PMID: 39259446 DOI: 10.1007/s12015-024-10788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
The discovery of adult neurogenesis in the middle of the past century is considered one of the most important breakthroughs in neuroscience. Despite its controversial nature, this discovery shaped our concept of neural plasticity, revolutionizing the way we look at our brains. In fact, after the discovery of adult neurogenesis, we started to consider the brain as something even more dynamic and highly adaptable. In neurogenic niches, adult neurogenesis is supported by neural stem cells (NSCs). These cells possess a unique set of characteristics such as being quiescent for long periods while actively sensing and reacting to their surroundings to influence a multitude of processes, including the generation of new neurons and glial cells. Therefore, NSCs can be viewed as sentinels to our brain's homeostasis, being able to replace damaged cells and simultaneously secrete numerous factors that restore regular brain function. In addition, it is becoming increasingly evident that NSCs play a central role in memory formation and consolidation. In this review, we will dissect how NSCs influence their surroundings through paracrine and autocrine types of action. We will also depict the mechanism of action of each factor. Finally, we will describe how NSCs integrate different and often opposing signals to guide their fate.
Collapse
Affiliation(s)
- João F Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
2
|
Reddy Onteddu V, Bhattacharya A, Baker NE. The Id protein Extramacrochaetae restrains the E protein Daughterless to regulate Notch, Rap1, and Sevenless within the R7 equivalence group of the Drosophila eye. Biol Open 2024; 13:bio060124. [PMID: 39041866 PMCID: PMC11360143 DOI: 10.1242/bio.060124] [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: 08/16/2023] [Accepted: 07/15/2024] [Indexed: 07/24/2024] Open
Abstract
The Drosophila Id gene extramacrochaetae (emc) is required during Drosophila eye development for proper cell fate specification within the R7 equivalence group. Without emc, R7 cells develop like R1/6 cells, and there are delays and deficits in differentiation of non-neuronal cone cells. Although emc encodes an Inhibitor of DNA-binding (Id) protein that is known to antagonize proneural bHLH protein function, no proneural gene is known for R7 or cone cell fates. These fates are also independent of daughterless (da), which encodes the ubiquitous E protein heterodimer partner of proneural bHLH proteins. We report here that the effects of emc mutations disappear in the absence of da, and are partially mimicked by forced expression of Da dimers, indicating that emc normally restrains da from interfering with R7 and cone cell specification, as occurs in emc mutants. emc, and da, regulate three known contributors to R7 fate, which are Notch signaling, Rap1, and Sevenless. R7 specification is partially restored to emc mutant cells by mutation of RapGap1, confirming that Rap1 activity, in addition to Notch activity, is a critical target of emc. These findings exemplify how mutations of an Id protein gene can affect processes that do not require any bHLH protein, by restraining Da activity within physiological bounds.
Collapse
Affiliation(s)
- Venkateswara Reddy Onteddu
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461,USA
| | - Abhishek Bhattacharya
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461,USA
| | - Nicholas E. Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461,USA
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461,USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461,USA
| |
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 affect 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. We found that multiple effects of emc mutations on cell growth and on eye development were all caused by reduced Diap1 levels and corresponding 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. 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 Neuroscience 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
|
Baloul S, Roussos C, Gomez-Lamarca M, Muresan L, Bray S. Changes in searching behaviour of CSL transcription complexes in Notch active conditions. Life Sci Alliance 2024; 7:e202302336. [PMID: 38097371 PMCID: PMC10721712 DOI: 10.26508/lsa.202302336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/03/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
During development cells receive a variety of signals, which are of crucial importance to their fate determination. One such source of signal is the Notch signalling pathway, where Notch activity regulates expression of target genes through the core transcription factor CSL. To understand changes in transcription factor behaviour that lead to transcriptional changes in Notch active cells, we have probed CSL behaviours in real time, using in vivo Single Molecule Localisation Microscopy. Trajectory analysis reveals that Notch-On conditions increase the fraction of bound CSL molecules, but also the proportion of molecules with exploratory behaviours. These properties are shared by the co-activator Mastermind. Furthermore, both CSL and Mastermind, exhibit characteristics of local exploration near a Notch target locus. A similar behaviour is observed for CSL molecules diffusing in the vicinity of other bound CSL clusters. We suggest therefore that CSL acquires an exploratory behaviour when part of the activation complex, favouring local searching and retention close to its target enhancers. This change explains how CSL can efficiently increase its occupancy at target sites in Notch-On conditions.
Collapse
Affiliation(s)
- Sarah Baloul
- https://ror.org/013meh722 Physiology Development and Neuroscience, Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | - Charalambos Roussos
- https://ror.org/013meh722 Physiology Development and Neuroscience, Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | - Maria Gomez-Lamarca
- https://ror.org/013meh722 Physiology Development and Neuroscience, Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | - Leila Muresan
- https://ror.org/013meh722 Physiology Development and Neuroscience, Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| | - Sarah Bray
- https://ror.org/013meh722 Physiology Development and Neuroscience, Cambridge Advanced Imaging Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
5
|
Mannino MC, Cassidy MB, Florez S, Rusan Z, Chakraborty S, Schoborg T. Mutations in abnormal spindle disrupt temporal transcription factor expression and trigger immune responses in the Drosophila brain. Genetics 2023; 225:iyad188. [PMID: 37831641 PMCID: PMC10697820 DOI: 10.1093/genetics/iyad188] [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: 08/30/2023] [Revised: 08/30/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The coordination of cellular behaviors during neurodevelopment is critical for determining the form, function, and size of the central nervous system (CNS). Mutations in the vertebrate Abnormal Spindle-Like, Microcephaly Associated (ASPM) gene and its Drosophila melanogaster ortholog abnormal spindle (asp) lead to microcephaly (MCPH), a reduction in overall brain size whose etiology remains poorly defined. Here, we provide the neurodevelopmental transcriptional landscape for a Drosophila model for autosomal recessive primary microcephaly-5 (MCPH5) and extend our findings into the functional realm to identify the key cellular mechanisms responsible for Asp-dependent brain growth and development. We identify multiple transcriptomic signatures, including new patterns of coexpressed genes in the developing CNS. Defects in optic lobe neurogenesis were detected in larval brains through downregulation of temporal transcription factors (tTFs) and Notch signaling targets, which correlated with a significant reduction in brain size and total cell numbers during the neurogenic window of development. We also found inflammation as a hallmark of asp mutant brains, detectable throughout every stage of CNS development, which also contributes to the brain size phenotype. Finally, we show that apoptosis is not a primary driver of the asp mutant brain phenotypes, further highlighting an intrinsic Asp-dependent neurogenesis promotion mechanism that is independent of cell death. Collectively, our results suggest that the etiology of the asp mutant brain phenotype is complex and that a comprehensive view of the cellular basis of the disorder requires an understanding of how multiple pathway inputs collectively determine tissue size and architecture.
Collapse
Affiliation(s)
- Maria C Mannino
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | | | - Steven Florez
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Zeid Rusan
- Personalis, Inc., Fremont, CA 94555, USA
| | - Shalini Chakraborty
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Todd Schoborg
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
6
|
He Q, Hou T, Fan X, Wang S, Wang Y, Chen S. Juvenile hormone suppresses sensory organ precursor determination to block Drosophila adult abdomen morphogenesis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103957. [PMID: 37192726 DOI: 10.1016/j.ibmb.2023.103957] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Juvenile hormone (JH) has a classic "status quo" action at both the pupal and adult molts when administrated exogenously. In Drosophila, treatment with JH at pupariation inhibits the formation of abdominal bristles, which are derived from the histoblasts. However, the mechanism via which JH exerts this effect remains poorly understood. In this study, we analyzed the effect of JH on histoblast proliferation, migration, and differentiation. Our results indicated that whereas the proliferation and migration of histoblasts remained unaffected following treatment with a JH mimic (JHM), their differentiation, particularly the specification of sensor organ precursor (SOP) cells, was inhibited. This effect was attributable to downregulated proneural genes achaete (ac) and Scute (sc) expression levels, which prevented the specification of SOP cells in proneural clusters. Moreover, Kr-h1 was found to mediate this effect of JHM. Histoblast-specific overexpression or knockdown of Kr-h1, respectively mimicked or attenuated the effects exerted by JHM on abdominal bristle formation, SOP determination, and transcriptional regulation of ac and sc. These results indicated that the defective SOP determination was responsible for the inhibition of abdominal bristle formation by JHM, which, in turn, was mainly mediated via the transducing action of Kr-h1.
Collapse
Affiliation(s)
- Qianyu He
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China.
| | - Tianlan Hou
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xiaochun Fan
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shunxin Wang
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yanhong Wang
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shanshan Chen
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
7
|
Mannino MC, Bartels Cassidy M, Florez S, Rusan Z, Chakraborty S, Schoborg T. The neurodevelopmental transcriptome of the Drosophila melanogaster microcephaly gene abnormal spindle reveals a role for temporal transcription factors and the immune system in regulating brain size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.523369. [PMID: 36711768 PMCID: PMC9882087 DOI: 10.1101/2023.01.09.523369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The coordination of cellular behaviors during neurodevelopment is critical for determining the form, function, and size of the central nervous system. Mutations in the vertebrate Abnormal Spindle-Like, Microcephaly Associated (ASPM) gene and its Drosophila melanogaster ortholog abnormal spindle (asp) lead to microcephaly, a reduction in overall brain size whose etiology remains poorly defined. Here we provide the neurodevelopmental transcriptional landscape for a Drosophila model for autosomal recessive primary microcephaly (MCPH) and extend our findings into the functional realm in an attempt to identify the key cellular mechanisms responsible for Asp-dependent brain growth and development. We identify multiple transcriptomic signatures, including new patterns of co-expressed genes in the developing CNS. Defects in optic lobe neurogenesis were detected in larval brains through downregulation of temporal transcription factors (tTFs) and Notch signaling targets, which correlated with a significant reduction in brain size and total cell numbers during the neurogenic window of development. We also found inflammation as a hallmark of asp MCPH brains, detectable throughout every stage of CNS development, which also contributes to the brain size phenotype. Finally, we show that apoptosis is not a primary driver of the asp MCPH phenotype, further highlighting an intrinsic Asp-dependent neurogenesis promotion mechanism that is independent of cell death. Collectively, our results suggest that the etiology of asp MCPH is complex and that a comprehensive view of the cellular basis of the disorder requires an understanding of how multiple pathway inputs collectively determine the microcephaly phenotype.
Collapse
Affiliation(s)
- Maria C. Mannino
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | | | - Steven Florez
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | | | - Shalini Chakraborty
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| | - Todd Schoborg
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
8
|
Abstract
Notch signalling is a well-conserved signalling pathway that regulates cell fate through cell-cell communication. A typical feature of Notch signalling is ‘lateral inhibition’, whereby two neighbouring cells of equivalent state of differentiation acquire different cell fates. Recently, mathematical and computational approaches have addressed the Notch dynamics in Drosophila neural development. Typical examples of lateral inhibition are observed in the specification of neural stem cells in the embryo and sensory organ precursors in the thorax. In eye disc development, Notch signalling cooperates with other signalling pathways to define the evenly spaced positioning of the photoreceptor cells. The interplay between Notch and epidermal growth factor receptor signalling regulates the timing of neural stem cell differentiation in the optic lobe. In this review, we summarize the theoretical studies that have been conducted to elucidate the Notch dynamics in these systems and discuss the advantages of combining mathematical models with biological experiments.
Collapse
Affiliation(s)
- Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan.,Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
9
|
Johnson RI. Hexagonal patterning of the Drosophila eye. Dev Biol 2021; 478:173-182. [PMID: 34245727 DOI: 10.1016/j.ydbio.2021.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 10/24/2022]
Abstract
A complex network of transcription factor interactions propagates across the larval eye disc to establish columns of evenly-spaced R8 precursor cells, the founding cells of Drosophila ommatidia. After the recruitment of additional photoreceptors to each ommatidium, the surrounding cells are organized into their stereotypical pattern during pupal development. These support cells - comprised of pigment and cone cells - are patterned to encapsulate the photoreceptors and separate ommatidia with an hexagonal honeycomb lattice. Since the proteins and processes essential for correct eye patterning are conserved, elucidating how these function and change during Drosophila eye patterning can substantially advance our understanding of transcription factor and signaling networks, cytoskeletal structures, adhesion complexes, and the biophysical properties of complex tissues during their morphogenesis. Our understanding of many of these aspects of Drosophila eye patterning is largely descriptive. Many important questions, especially relating to the regulation and integration of cellular events, remain.
Collapse
Affiliation(s)
- Ruth I Johnson
- Biology Department, Wesleyan University, 52 Lawn Avenue, Middletown, CT, USA.
| |
Collapse
|
10
|
DeAngelis MW, Coolon JD, Johnson RI. Comparative transcriptome analyses of the Drosophila pupal eye. G3-GENES GENOMES GENETICS 2021; 11:5995320. [PMID: 33561221 PMCID: PMC8043229 DOI: 10.1093/g3journal/jkaa003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/08/2020] [Indexed: 12/04/2022]
Abstract
Tissue function is dependent on correct cellular organization and behavior. As a result, the identification and study of genes that contribute to tissue morphogenesis is of paramount importance to the fields of cell and developmental biology. Many of the genes required for tissue patterning and organization are highly conserved between phyla. This has led to the emergence of several model organisms and developmental systems that are used to study tissue morphogenesis. One such model is the Drosophila melanogaster pupal eye that has a highly stereotyped arrangement of cells. In addition, the pupal eye is postmitotic that allows for the study of tissue morphogenesis independent from any effects of proliferation. While the changes in cell morphology and organization that occur throughout pupal eye development are well documented, less is known about the corresponding transcriptional changes that choreograph these processes. To identify these transcriptional changes, we dissected wild-type Canton S pupal eyes and performed RNA-sequencing. Our analyses identified differential expression of many loci that are documented regulators of pupal eye morphogenesis and contribute to multiple biological processes including signaling, axon projection, adhesion, and cell survival. We also identified differential expression of genes not previously implicated in pupal eye morphogenesis such as components of the Toll pathway, several non-classical cadherins, and components of the muscle sarcomere, which could suggest these loci function as novel patterning factors.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Joseph D Coolon
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Ruth I Johnson
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| |
Collapse
|
11
|
Willink B, Duryea MC, Wheat C, Svensson EI. Changes in gene expression during female reproductive development in a color polymorphic insect. Evolution 2020; 74:1063-1081. [DOI: 10.1111/evo.13979] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 03/19/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Beatriz Willink
- Department of Biology, Evolutionary Ecology Unit, Ecology BuildingLund University Lund 223–62 Sweden
- Current Address: School of BiologyUniversity of Costa Rica San José 11501–2060 Costa Rica
| | | | | | - Erik I. Svensson
- Department of Biology, Evolutionary Ecology Unit, Ecology BuildingLund University Lund 223–62 Sweden
| |
Collapse
|
12
|
Couturier L, Mazouni K, Corson F, Schweisguth F. Regulation of Notch output dynamics via specific E(spl)-HLH factors during bristle patterning in Drosophila. Nat Commun 2019; 10:3486. [PMID: 31375669 PMCID: PMC6677740 DOI: 10.1038/s41467-019-11477-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/03/2019] [Indexed: 12/13/2022] Open
Abstract
The stereotyped arrangement of sensory bristles on the adult fly thorax arises from a self-organized process, in which inhibitory Notch signaling both delimits proneural stripes and singles out sensory organ precursor cells (SOPs). A dynamic balance between proneural factors and Enhancer of split-HLH (E(spl)-HLH) Notch targets underlies patterning, but how this is regulated is unclear. Here, were identify two classes of E(spl)-HLH factors, whose expression both precedes and delimits proneural activity, and is dependent on proneural activity and required for proper SOP spacing within the stripes, respectively. These two classes are partially redundant, since a member of the second class, that is normally cross-repressed by members of the first class, can functionally compensate for their absence. The regulation of specific E(spl)-HLH genes by proneural factors amplifies the response to Notch as SOPs are being selected, contributing to patterning dynamics in the notum, and likely operates in other developmental contexts. The patterning of sensory bristles on the dorsal thorax of flies is regulated by two transcription factor families but the dynamics of this regulation is unclear. Here, the authors visualize seven E(spl)-HLH proteins, showing their regulated expression promotes mutual inhibition by Notch during notum patterning.
Collapse
Affiliation(s)
- Lydie Couturier
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015, Paris, France.,CNRS, UMR3738, 75015, Paris, France
| | - Khalil Mazouni
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015, Paris, France.,CNRS, UMR3738, 75015, Paris, France
| | - Francis Corson
- Laboratoire de Physique de l'Ecole Normale Supérieure, CNRS, Sorbonne Université, Université Paris Diderot, 75005, Paris, France
| | - François Schweisguth
- Department of Developmental and Stem Cell Biology, Institut Pasteur, 75015, Paris, France. .,CNRS, UMR3738, 75015, Paris, France.
| |
Collapse
|
13
|
Xu X, Zhu H, Yang F, Wu C, Jiang C, Yu W, Liu K, Sheng Q, Nie Z. Bmo-miR-79 downregulates the expression of BmEm4 in the silkworm, Bombyx mori. Gene 2019; 690:113-119. [PMID: 30593917 DOI: 10.1016/j.gene.2018.12.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/30/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022]
Abstract
MicroRNA is an important regulation factor in insect development and metamorphosis. It has been reported that E(spl)m4 is a miRNA-targeted gene, as well as the target of the Notch signaling pathway in Drosophila. The expression of E(spl)m4 can be regulated by microRNA and further affect the neural development of Drosophila. Here, we found that BmEm4, an ortholog of E(spl)m4 from Bombyx mori, was the target gene of bmo-miR-79, with target sites containing the Brd and K boxes of the BmEm4_3'UTR, which was validated by the dual luciferase reporter (DLR) assay. Furthermore, bmo-miR-79 mimics can inhibit the expression of BmEm4 in BmN cells after transfection, and bmo-miR-79 can also inhibit the expression of BmEm4 in different developmental stages of Bombyx mori at a posttranscriptional level, to different degrees. The EMSA test further showed that bmo-miR-79 could bind to BmAGO2, which is the Bombyx mori argonaute2 protein, suggesting that bmo-miR-79 might regulate the expression of BmEm4 by forming miRISC complexes with BmAGO2. Taken together, bmo-miR-79 could regulate the expression of BmEm4 mediated by BmAGO2 and further affect its function in the silkworm Bombyx mori.
Collapse
Affiliation(s)
- Xiaoyuan Xu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Honglin Zhu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Fan Yang
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chengcheng Wu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Caiying Jiang
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Wei Yu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Kuancheng Liu
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Qing Sheng
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zuoming Nie
- College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
14
|
Salazar JL, Yamamoto S. Integration of Drosophila and Human Genetics to Understand Notch Signaling Related Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:141-185. [PMID: 30030826 PMCID: PMC6233323 DOI: 10.1007/978-3-319-89512-3_8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Notch signaling research dates back to more than one hundred years, beginning with the identification of the Notch mutant in the fruit fly Drosophila melanogaster. Since then, research on Notch and related genes in flies has laid the foundation of what we now know as the Notch signaling pathway. In the 1990s, basic biological and biochemical studies of Notch signaling components in mammalian systems, as well as identification of rare mutations in Notch signaling pathway genes in human patients with rare Mendelian diseases or cancer, increased the significance of this pathway in human biology and medicine. In the 21st century, Drosophila and other genetic model organisms continue to play a leading role in understanding basic Notch biology. Furthermore, these model organisms can be used in a translational manner to study underlying mechanisms of Notch-related human diseases and to investigate the function of novel disease associated genes and variants. In this chapter, we first briefly review the major contributions of Drosophila to Notch signaling research, discussing the similarities and differences between the fly and human pathways. Next, we introduce several biological contexts in Drosophila in which Notch signaling has been extensively characterized. Finally, we discuss a number of genetic diseases caused by mutations in genes in the Notch signaling pathway in humans and we expand on how Drosophila can be used to study rare genetic variants associated with these and novel disorders. By combining modern genomics and state-of-the art technologies, Drosophila research is continuing to reveal exciting biology that sheds light onto mechanisms of disease.
Collapse
Affiliation(s)
- Jose L Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX, USA.
- Program in Developmental Biology, BCM, Houston, TX, USA.
- Department of Neuroscience, BCM, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
15
|
Wu C, Jordan MD, Newcomb RD, Gemmell NJ, Bank S, Meusemann K, Dearden PK, Duncan EJ, Grosser S, Rutherford K, Gardner PP, Crowhurst RN, Steinwender B, Tooman LK, Stevens MI, Buckley TR. Analysis of the genome of the New Zealand giant collembolan (Holacanthella duospinosa) sheds light on hexapod evolution. BMC Genomics 2017; 18:795. [PMID: 29041914 PMCID: PMC5644144 DOI: 10.1186/s12864-017-4197-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/08/2017] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The New Zealand collembolan genus Holacanthella contains the largest species of springtails (Collembola) in the world. Using Illumina technology we have sequenced and assembled a draft genome and transcriptome from Holacanthella duospinosa (Salmon). We have used this annotated assembly to investigate the genetic basis of a range of traits critical to the evolution of the Hexapoda, the phylogenetic position of H. duospinosa and potential horizontal gene transfer events. RESULTS Our genome assembly was ~375 Mbp in size with a scaffold N50 of ~230 Kbp and sequencing coverage of ~180×. DNA elements, LTRs and simple repeats and LINEs formed the largest components and SINEs were very rare. Phylogenomics (370,877 amino acids) placed H. duospinosa within the Neanuridae. We recovered orthologs of the conserved sex determination genes thought to play a role in sex determination. Analysis of CpG content suggested the absence of DNA methylation, and consistent with this we were unable to detect orthologs of the DNA methyltransferase enzymes. The small subunit rRNA gene contained a possible retrotransposon. The Hox gene complex was broken over two scaffolds. For chemosensory ability, at least 15 and 18 ionotropic glutamate and gustatory receptors were identified, respectively. However, we were unable to identify any odorant receptors or their obligate co-receptor Orco. Twenty-three chitinase-like genes were identified from the assembly. Members of this multigene family may play roles in the digestion of fungal cell walls, a common food source for these saproxylic organisms. We also detected 59 and 96 genes that blasted to bacteria and fungi, respectively, but were located on scaffolds that otherwise contained arthropod genes. CONCLUSIONS The genome of H. duospinosa contains some unusual features including a Hox complex broken over two scaffolds, in a different manner to other arthropod species, a lack of odorant receptor genes and an apparent lack of environmentally responsive DNA methylation, unlike many other arthropods. Our detection of candidate horizontal gene transfer candidates confirms that this phenomenon is occurring across Collembola. These findings allow us to narrow down the regions of the arthropod phylogeny where key innovations have occurred that have facilitated the evolutionary success of Hexapoda.
Collapse
Affiliation(s)
- Chen Wu
- Landcare Research, Private Bag, Auckland, 92170, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Melissa D Jordan
- The New Zealand Institute for Plant & Food Research Ltd, Auckland, New Zealand
| | - Richard D Newcomb
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- The New Zealand Institute for Plant & Food Research Ltd, Auckland, New Zealand
| | - Neil J Gemmell
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sarah Bank
- Center for Molecular Biodiversity Research, Zoological Research Museum Alexander Koenig, Adenauerallee 160, 53113, Bonn, Germany
| | - Karen Meusemann
- Center for Molecular Biodiversity Research, Zoological Research Museum Alexander Koenig, Adenauerallee 160, 53113, Bonn, Germany
- Evolutionary Biology & Ecology, Institute for Biology, University of Freiburg, Freiburg, Germany
| | - Peter K Dearden
- Genetics Otago, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Elizabeth J Duncan
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Sefanie Grosser
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Animal Behaviour, Bielefeld University, Bielefeld, Germany
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilian University of Munich, Planegg-, Martinsried, Germany
| | - Kim Rutherford
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Paul P Gardner
- Biomolecular Interactions Centre, School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Ross N Crowhurst
- The New Zealand Institute for Plant & Food Research Ltd, Auckland, New Zealand
| | - Bernd Steinwender
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- The New Zealand Institute for Plant & Food Research Ltd, Auckland, New Zealand
| | - Leah K Tooman
- The New Zealand Institute for Plant & Food Research Ltd, Auckland, New Zealand
| | - Mark I Stevens
- South Australian Museum, North Terrace, GPO Box 234, Adelaide, SA, 5001, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Thomas R Buckley
- Landcare Research, Private Bag, Auckland, 92170, New Zealand.
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
16
|
Majot AT, Bidwai AP. Analysis of transient hypermorphic activity of E(spl)D during R8 specification. PLoS One 2017; 12:e0186439. [PMID: 29036187 PMCID: PMC5643056 DOI: 10.1371/journal.pone.0186439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/02/2017] [Indexed: 11/23/2022] Open
Abstract
Drosophila atonal (ato) is required for the specification of founding R8 photoreceptors during retinal development. ato is regulated via dual eye-specific enhancers; ato-3’ is subject to initial induction whereas 5’-ato facilitates Notch-mediated autoregulation. Notch is further utilized to induce bHLH repressors of the E(spl) locus to restrict Ato from its initial broad expression to individual cells. Although Notch operates in two, distinct phases, it has remained unclear how the two phases maintain independence from one another. The difference in these two phases has attributed to the hypothesized delayed expression of E(spl). However, immunofluorescence data indicate that E(spl) are expressed during early Ato patterning, suggesting a more sophisticated underlying mechanism. To probe this mechanism, we provide evidence that although E(spl) exert no influence on ato-3’, E(spl) repress 5’-ato and deletion of the E(spl) locus elicits precocious 5’-ato activity. Thus, E(spl) imposes a delay to the timing in which Ato initiates autoregulation. We next sought to understand this finding in the context of E(spl)D, which encodes a dysregulated variant of E(spl)M8 that perturbs R8 patterning, though, as previously reported, only in conjunction with the mutant receptor Nspl. We established a genetic interaction between E(spl)D and roughened eye (roe), a known modulator of Notch signaling in retinogenesis. This link further suggests a dosage-dependence between E(spl) and the proneural activators Ato and Sens, as indicated via interaction assays in which E(spl)D renders aberrant R8 patterning in conjunction with reduced proneural dosage. In total, the biphasicity of Notch signaling relies, to some degree, on the post-translational regulation of individual E(spl) members and, importantly, that post-translational regulation is likely necessary to modulate the level of E(spl) activity throughout the progression of Ato expression.
Collapse
Affiliation(s)
- Adam T. Majot
- Department of Biology, West Virginia University, Morgantown, West Virginia, United States of America
| | - Ashok P. Bidwai
- Department of Biology, West Virginia University, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
17
|
Dey NS, Ramesh P, Chugh M, Mandal S, Mandal L. Dpp dependent Hematopoietic stem cells give rise to Hh dependent blood progenitors in larval lymph gland of Drosophila. eLife 2016; 5:18295. [PMID: 27782877 PMCID: PMC5120881 DOI: 10.7554/elife.18295] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 10/25/2016] [Indexed: 12/19/2022] Open
Abstract
Drosophila hematopoiesis bears striking resemblance with that of vertebrates, both in the context of distinct phases and the signaling molecules. Even though, there has been no evidence of Hematopoietic stem cells (HSCs) in Drosophila, the larval lymph gland with its Hedgehog dependent progenitors served as an invertebrate model of progenitor biology. Employing lineage-tracing analyses, we have now identified Notch expressing HSCs in the first instar larval lymph gland. Our studies clearly establish the hierarchical relationship between Notch expressing HSCs and the previously described Domeless expressing progenitors. These HSCs require Decapentapelagic (Dpp) signal from the hematopoietic niche for their maintenance in an identical manner to vertebrate aorta-gonadal-mesonephros (AGM) HSCs. Thus, this study not only extends the conservation across these divergent taxa, but also provides a new model that can be exploited to gain better insight into the AGM related Hematopoietic stem cells (HSCs).
Collapse
Affiliation(s)
- Nidhi Sharma Dey
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Parvathy Ramesh
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Mayank Chugh
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
18
|
Gunnar E, Bivik C, Starkenberg A, Thor S. sequoia controls the type I>0 daughter proliferation switch in the developing Drosophila nervous system. Development 2016; 143:3774-3784. [PMID: 27578794 DOI: 10.1242/dev.139998] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/22/2016] [Indexed: 01/16/2023]
Abstract
Neural progenitors typically divide asymmetrically to renew themselves, while producing daughters with more limited potential. In the Drosophila embryonic ventral nerve cord, neuroblasts initially produce daughters that divide once to generate two neurons/glia (type I proliferation mode). Subsequently, many neuroblasts switch to generating daughters that differentiate directly (type 0). This programmed type I>0 switch is controlled by Notch signaling, triggered at a distinct point of lineage progression in each neuroblast. However, how Notch signaling onset is gated was unclear. We recently identified Sequoia (Seq), a C2H2 zinc-finger transcription factor with homology to Drosophila Tramtrack (Ttk) and the positive regulatory domain (PRDM) family, as important for lineage progression. Here, we find that seq mutants fail to execute the type I>0 daughter proliferation switch and also display increased neuroblast proliferation. Genetic interaction studies reveal that seq interacts with the Notch pathway, and seq furthermore affects expression of a Notch pathway reporter. These findings suggest that seq may act as a context-dependent regulator of Notch signaling, and underscore the growing connection between Seq, Ttk, the PRDM family and Notch signaling.
Collapse
Affiliation(s)
- Erika Gunnar
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping SE-58185, Sweden
| | - Caroline Bivik
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping SE-58185, Sweden
| | - Annika Starkenberg
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping SE-58185, Sweden
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, Linkoping SE-58185, Sweden
| |
Collapse
|
19
|
Duncan EJ, Hyink O, Dearden PK. Notch signalling mediates reproductive constraint in the adult worker honeybee. Nat Commun 2016; 7:12427. [PMID: 27485026 PMCID: PMC4976197 DOI: 10.1038/ncomms12427] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
The hallmark of eusociality is the reproductive division of labour, in which one female caste reproduces, while reproduction is constrained in the subordinate caste. In adult worker honeybees (Apis mellifera) reproductive constraint is conditional: in the absence of the queen and brood, adult worker honeybees activate their ovaries and lay haploid male eggs. Here, we demonstrate that chemical inhibition of Notch signalling can overcome the repressive effect of queen pheromone and promote ovary activity in adult worker honeybees. We show that Notch signalling acts on the earliest stages of oogenesis and that the removal of the queen corresponds with a loss of Notch protein in the germarium. We conclude that the ancient and pleiotropic Notch signalling pathway has been co-opted into constraining reproduction in worker honeybees and we provide the first molecular mechanism directly linking ovary activity in adult worker bees with the presence of the queen. In honeybees, pheromones produced by the queen inhibit reproduction by workers and enforce a eusocial division of labour. Here, Duncan, Hyink and Dearden show that this inhibition is mediated by the Notch signalling pathway in the workers' ovaries.
Collapse
Affiliation(s)
- Elizabeth J Duncan
- Department of Biochemistry, Laboratory for Evolution and Development, Genetics Otago and Gravida (The National Centre for Growth and Development), University of Otago, P.O. Box 56, Dunedin 9054, Aotearoa-New Zealand
| | - Otto Hyink
- Department of Biochemistry, Laboratory for Evolution and Development, Genetics Otago and Gravida (The National Centre for Growth and Development), University of Otago, P.O. Box 56, Dunedin 9054, Aotearoa-New Zealand
| | - Peter K Dearden
- Department of Biochemistry, Laboratory for Evolution and Development, Genetics Otago and Gravida (The National Centre for Growth and Development), University of Otago, P.O. Box 56, Dunedin 9054, Aotearoa-New Zealand
| |
Collapse
|
20
|
Zhang T, Du W. Groucho restricts rhomboid expression and couples EGFR activation with R8 selection during Drosophila photoreceptor differentiation. Dev Biol 2015; 407:246-55. [PMID: 26417727 DOI: 10.1016/j.ydbio.2015.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 07/18/2015] [Accepted: 09/24/2015] [Indexed: 11/17/2022]
Abstract
Notch and EGFR signaling pathways play important roles in photoreceptor differentiation during Drosophila eye development. Notch signaling induces Enhancer of Split (E(spl)) proteins to repress atonal (ato) expression and restrict R8 photoreceptor cell fate. The R8 precursors express rhomboid (rho), which is required for the release of active EGFR ligand to activate EGFR signaling in surrounding cells for the subsequent stepwise recruitment. However, it is not clear about the mechanisms of transcriptional regulation of rho and how the lateral inhibition of Notch signaling and rho expression are coordinated. In this study, we show that inactivation of Groucho (Gro), an evolutionally conserved transcriptional corepressor, inhibits Ato upregulation, delays R8 determination, and promotes differentiation of R2-5 type of neurons. We demonstrate that these phenotypes are caused by a combination of the loss of Notch-mediated lateral inhibition and the precocious activation of EGFR signaling due to deregulated rho expression. Blocking EGFR signaling by Pnt-RNAi in conjunction with Gro-inactivation leads to lateral inhibition defects with deregulated Ato expression and R8 differentiation. We further show that inactivation of E(spl), which are the Gro binding transcription factors, causes deregulated rho expression and extra R8 cells within and posterior to the morphogenetic furrow (MF), and that E(spl) mediates the binding of Gro to the regulatory regions of both rho and ato genes in eye disc cells. Our results suggest that Gro inhibits rho expression in undifferentiated cells and represses the expression of both ato and rho in non-R8 precursors during initiation of photoreceptor differentiation in an E(spl)-dependent manner. The latter function of Gro provides novel insights into the mechanism that coordinates R8 specification with the restriction of initial rho expression to developing R8 cells.
Collapse
Affiliation(s)
- Tianyi Zhang
- Ben May Department for Cancer Research, The University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA
| | - Wei Du
- Ben May Department for Cancer Research, The University of Chicago, 929 E. 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
21
|
Dearden PK. Origin and evolution of the enhancer of split complex. BMC Genomics 2015; 16:712. [PMID: 26384649 PMCID: PMC4575448 DOI: 10.1186/s12864-015-1926-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/12/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The Enhancer of split complex is an unusual gene complex found in Arthropod genomes. Where known this complex of genes is often regulated by Notch cell signalling and is critically important for neurogenesis. The Enhancer of split complex is made up of two different classes of genes, basic helix-loop-helix-orange domain transcription factors and bearded class genes. The association of these genes has been detected in the genomes of insects and crustaceans. RESULTS Tracing the evolution of the Enhancer of split complex in recently sequenced Arthropod genomes indicates that enhancer of split basic helix-loop-helix orange domain genes arose before the common ancestor of insects and Crustacea, and before the formation of the complex. Throughout insect and crustacean evolution, a four-gene cluster has been present with lineage specific gene losses and duplications. The complex can be found in the vast majority of genomes, but appears to be missing from the genomes of chalcid wasps, raising questions as to how they carry out neurogenesis in the absence of these crucial genes. CONCLUSIONS The enhancer of split complex arose in the common ancestor of Crustacea and insects, probably through the linkage of a basic helix-loop-helix orange domain gene and a bearded class gene. The complex has been maintained, with variations, throughout insect and crustacean evolution indicating some function of the complex, such as coordinate regulation, may maintain its structure through evolutionary time.
Collapse
Affiliation(s)
- Peter K Dearden
- Genetics Otago and Gravida (National Centre for Growth and Development), Biochemistry Department, University of Otago, Dunedin, Aotearoa, New Zealand.
| |
Collapse
|
22
|
Linneweber GA, Winking M, Fischbach KF. The Cell Adhesion Molecules Roughest, Hibris, Kin of Irre and Sticks and Stones Are Required for Long Range Spacing of the Drosophila Wing Disc Sensory Sensilla. PLoS One 2015; 10:e0128490. [PMID: 26053791 PMCID: PMC4459997 DOI: 10.1371/journal.pone.0128490] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/27/2015] [Indexed: 12/18/2022] Open
Abstract
Most animal tissues and organ systems are comprised of highly ordered arrays of varying cell types. The development of external sensory organs requires complex cell-cell communication in order to give each cell a specific identity and to ensure a regular distributed pattern of the sensory bristles. This involves both long and short range signaling mediated by either diffusible or cell anchored factors. In a variety of processes the heterophilic Irre Cell Recognition Module, consisting of the Neph-like proteins: Roughest, Kin of irre and of the Nephrin-like proteins: Sticks and Stones, Hibris, plays key roles in the recognition events of different cell types throughout development. In the present study these proteins are apically expressed in the adhesive belt of epithelial cells participating in sense organ development in a partially exclusive and asymmetric manner. Using mutant analysis the GAL4/UAS system, RNAi and gain of function we found an involvement of all four Irre Cell Recognition Module-proteins in the development of a highly structured array of sensory organs in the wing disc. The proteins secure the regular spacing of sensory organs showing partial redundancy and may function in early lateral inhibition events as well as in cell sorting processes. Comparisons with other systems suggest that the Irre Cell Recognition module is a key organizer of highly repetitive structures.
Collapse
Affiliation(s)
- Gerit Arne Linneweber
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Mathis Winking
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| | - Karl-Friedrich Fischbach
- Department of Neurobiology, Albert-Ludwigs-University Freiburg, Schänzlestr. 1, D-79104, Freiburg, Germany
| |
Collapse
|
23
|
Wurmbach E, Preiss A. Deletion mapping in the Enhancer of split complex. Hereditas 2015; 151:159-68. [PMID: 25588303 DOI: 10.1111/hrd2.00065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/26/2014] [Indexed: 11/30/2022] Open
Abstract
The Enhancer of split complex [E(spl)-C] comprises twelve genes of different classes. Seven genes encode proteins of with a basic-helix-loop-helix-orange (bHLH-O) domain that function as transcriptional repressors and serve as effectors of the Notch signalling pathway. They have been named E(spl)m8-, m7-, m5-, m3-, mβ-, mγ- and mδ-HLH. Four genes, E(spl)m6-, m4-, m2- and mα-BFM are intermingled and encode Notch repressor proteins of the Bearded-family (BFM). The complex is split by a single gene of unrelated function, encoding a Kazal-type protease inhibitor (Kaz-m1). All members within a family, bHLH-O or BFM, are very similar in structure and in function. In an attempt to generate specific mutants, we have mobilised P-element constructs residing next to E(spl)m7-HLH and E(spl)mγ-HLH, respectively. The resulting deletions were mapped molecularly and by cytology. Two small deletions affected only E(spl)m7-HLH and E(spl)mδ. The deficient flies were viable without apparent phenotype. Larger deletions, generated also by X-ray mutagenesis, uncover most of the E(spl)-C. The phenotypes of homozygous deficient embryos were analysed to characterize the respective loss of Notch signalling activity.
Collapse
Affiliation(s)
- Elisa Wurmbach
- Office of Chief Medical Examiner, Department of Forensic Biology, New York, NY, USA
| | | |
Collapse
|
24
|
Lu Y, Li Z. Notch signaling downstream target E(spl)mbeta is dispensable for adult midgut homeostasis in Drosophila. Gene 2015; 560:89-95. [PMID: 25637572 DOI: 10.1016/j.gene.2015.01.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/11/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
Abstract
Adult tissue homeostasis is maintained by residential stem cells through the proper balance of stem cell self-renewal and differentiation. The adult midgut of Drosophila contains multipotent intestinal stem cells (ISCs), and Notch signaling plays critical roles in the proliferation and differentiation of these ISCs. However, how Notch signaling downstream targets regulate ISC proliferation and differentiation still remains unclear. Here we find that Notch signaling downstream targets E(spl)mbeta and E(spl)malpha are differentially expressed in ISCs and their progeny. Interestingly, we find that midgut homeostasis is not affected in E(spl)mbeta null mutant. No obvious defects are observed in the intestines ectopically expressing E(spl)mbeta or E(spl)malpha. Importantly, we find that the defects in ISC proliferation and differentiation observed in Notch mutant cannot be rescued by ectopic expression of E(spl)mbeta or E(spl)malpha. Together, these data indicate that the proliferation and differentiation of ISCs are not regulated by individual Notch downstream target, but by different downstream targets collectively.
Collapse
Affiliation(s)
- Yanfen Lu
- Plant Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; Beijing Key Laboratory of New Technology in Agriculture Application, Beijing University of Agriculture, Beijing 102206, China.
| | - Zhouhua Li
- School of Life Sciences, Capital Normal University, Beijing 100048, China.
| |
Collapse
|
25
|
Abstract
The ability to visualize Notch pathway activity in vivo is invaluable for studying the functions and mechanisms of Notch signaling. A variety of tools have been developed to enable monitoring of pathway activity in Drosophila, including endogenous Notch-responsive genes and synthetic transcriptional reporter constructs. Here we summarize some of the different Notch signaling reporters that are available, discuss their relative merits, and describe two methods for visualizing their expression (immunostaining and X-gal staining). These approaches are widely applicable to a range of tissues and stages in Drosophila development.
Collapse
|
26
|
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: 15] [Impact Index Per Article: 1.7] [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
|
27
|
Neural precursor-specific expression of multiple Drosophila genes is driven by dual enhancer modules with overlapping function. Proc Natl Acad Sci U S A 2014; 111:17194-9. [PMID: 25404315 DOI: 10.1073/pnas.1415308111] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Transcriptional cis-regulatory modules (CRMs), or enhancers, are responsible for directing gene expression in specific territories and cell types during development. In some instances, the same gene may be served by two or more enhancers with similar specificities. Here we show that the utilization of dual, or "shadow", enhancers is a common feature of genes that are active specifically in neural precursor (NP) cells in Drosophila. By genome-wide computational discovery of statistically significant clusters of binding motifs for both proneural activator (P) proteins and basic helix-loop-helix (bHLH) repressor (R) factors (a "P+R" regulatory code), we have identified NP-specific enhancer modules associated with multiple genes expressed in this cell type. These CRMs are distinct from those previously identified for the corresponding gene, establishing the existence of a dual-enhancer arrangement in which both modules reside close to the gene they serve. Using wild-type and mutant reporter gene constructs in vivo, we show that P sites in these modules mediate activation by proneural factors in "proneural cluster" territories, whereas R sites mediate repression by bHLH repressors, which serves to restrict expression specifically to NP cells. To our knowledge, our results identify the first direct targets of these bHLH repressors. Finally, using genomic rescue constructs for neuralized (neur), we demonstrate that each of the gene's two NP-specific enhancers is sufficient to rescue neur function in the lateral inhibition process by which adult sensory organ precursor (SOP) cells are specified, but that deletion of both enhancers results in failure of this event.
Collapse
|
28
|
Nagel AC, Preiss A. Mutation of potential MAPK phosphorylation sites in the Notch antagonist Hairless. Hereditas 2014; 151:102-8. [PMID: 25363277 DOI: 10.1111/hrd2.00066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/31/2014] [Indexed: 01/05/2023] Open
Abstract
Cellular differentiation during eumetazoan development is based on highly conserved signalling pathways. Two of them, the Notch and the EGFR signalling pathways, are closely intertwined. We have identified two potential target sites of the Mitogen activated kinase (MAPK), the downstream effector kinase of EGFR, within Hairless (H), the major antagonist of Notch signalling in Drosophila. Assuming that phosphorylation of these sites modulates H activity, a direct influence of EGFR signalling on Notch pathway regulation might be possible. This hypothesis was tested by generating a phospho-deficient and a phospho-mimetic H isoform and by assaying for their biological activity. We first addressed the binding of known H interaction partners Su(H), Gro, CtBP and Pros26.4 which was similar between mutant and wild type H. Next we assayed eye, wing and bristle development which are strongly affected by the overexpression of H due to the inhibition of Notch signalling. Overexpression of the mutant constructs resulted in phenotypes similar to wildtype H overexpression, yet with subtle differences in phenotypic severity. However, large variations suggest that the mutated residues may be critical for the overall structure or stability of H. Albeit of minor impact, EGFR may fine tune Notch signalling via MAPK dependent phosphorylation of H.
Collapse
Affiliation(s)
- Anja C Nagel
- Universität Hohenheim, Institut für Genetik (240), Garbenstr. 30, DE-70599, Stuttgart, Germany.
| | | |
Collapse
|
29
|
Kaul A, Schuster E, Jennings BH. The Groucho co-repressor is primarily recruited to local target sites in active chromatin to attenuate transcription. PLoS Genet 2014; 10:e1004595. [PMID: 25165826 PMCID: PMC4148212 DOI: 10.1371/journal.pgen.1004595] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/03/2014] [Indexed: 12/25/2022] Open
Abstract
Gene expression is regulated by the complex interaction between transcriptional activators and repressors, which function in part by recruiting histone-modifying enzymes to control accessibility of DNA to RNA polymerase. The evolutionarily conserved family of Groucho/Transducin-Like Enhancer of split (Gro/TLE) proteins act as co-repressors for numerous transcription factors. Gro/TLE proteins act in several key pathways during development (including Notch and Wnt signaling), and are implicated in the pathogenesis of several human cancers. Gro/TLE proteins form oligomers and it has been proposed that their ability to exert long-range repression on target genes involves oligomerization over broad regions of chromatin. However, analysis of an endogenous gro mutation in Drosophila revealed that oligomerization of Gro is not always obligatory for repression in vivo. We have used chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq) to profile Gro recruitment in two Drosophila cell lines. We find that Gro predominantly binds at discrete peaks (<1 kilobase). We also demonstrate that blocking Gro oligomerization does not reduce peak width as would be expected if Gro oligomerization induced spreading along the chromatin from the site of recruitment. Gro recruitment is enriched in “active” chromatin containing developmentally regulated genes. However, Gro binding is associated with local regions containing hypoacetylated histones H3 and H4, which is indicative of chromatin that is not fully open for efficient transcription. We also find that peaks of Gro binding frequently overlap the transcription start sites of expressed genes that exhibit strong RNA polymerase pausing and that depletion of Gro leads to release of polymerase pausing and increased transcription at a bona fide target gene. Our results demonstrate that Gro is recruited to local sites by transcription factors to attenuate rather than silence gene expression by promoting histone deacetylation and polymerase pausing. Repression by transcription factors plays a central role in gene regulation. The Groucho/Transducin-Like Enhancer of split (Gro/TLE) family of co-repressors interacts with many different transcription factors and has many essential roles during animal development. Groucho/TLE proteins form oligomers that are necessary for target gene repression in some contexts. We have profiled the genome-wide recruitment of the founding member of this family, Groucho (from Drosophila) to gain insight into how and where it binds with respect to target genes and to identify factors associated with its binding. We find that Groucho binds in discrete peaks, frequently at transcription start sites, and that blocking Groucho from forming oligomers does not significantly change the pattern of Groucho recruitment. Although Groucho acts as a repressor, Groucho binding is enriched in chromatin that is permissive for transcription, and we find that it acts to attenuate rather than completely silence target gene expression. Thus, Groucho does not act as an “on/off” switch on target gene expression, but rather as a “mute” button.
Collapse
Affiliation(s)
- Aamna Kaul
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Eugene Schuster
- Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Barbara H. Jennings
- UCL Cancer Institute, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
30
|
Fernandes VM, Panchapakesan SSS, Braid LR, Verheyen EM. Nemo promotes Notch-mediated lateral inhibition downstream of proneural factors. Dev Biol 2014; 392:334-43. [PMID: 24880113 DOI: 10.1016/j.ydbio.2014.05.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 04/30/2014] [Accepted: 05/15/2014] [Indexed: 12/27/2022]
Abstract
During neurogenesis, conserved tissue-specific proneural factors establish a cell's competence to take on neural fate from within a field of unspecified cells. Proneural genes encode basic helix-loop-helix transcription factors that promote the expression of 'core' and subtype-specific target genes. Target genes include both pan-neuronal genes and genes that aid in the process of refinement, known as lateral inhibition. In this process, proneural gene expression is increased in the neural progenitor while simultaneously down-regulated in the surrounding cells, in a Notch signalling-dependent manner. Here, we identify nemo (nmo) as a target of members of both Drosophila Atonal and Achaete-Scute proneural factor families and find that mammalian proneural homologs induce Nemo-like-kinase (Nlk) expression in cell culture. We find that nmo loss of function leads to reduced expression of Notch targets and to perturbations in Notch-mediated lateral inhibition. Furthermore, Notch hyperactivity can compensate for nmo loss in the Drosophila eye. Thus nmo promotes Notch-mediated lateral inhibition downstream of proneural factors during neurogenesis.
Collapse
Affiliation(s)
- Vilaiwan M Fernandes
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada V5A1S6
| | - Shanker S S Panchapakesan
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada V5A1S6
| | - Lorena R Braid
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada V5A1S6; Defence Research and Development Canada - Suffield, Biotechnology Section, Medicine Hat, AB, Canada T1A 8K6
| | - Esther M Verheyen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada V5A1S6.
| |
Collapse
|
31
|
The Ser/Thr phosphatase PP2A regulatory subunit widerborst inhibits notch signaling. PLoS One 2014; 9:e101884. [PMID: 25006677 PMCID: PMC4090204 DOI: 10.1371/journal.pone.0101884] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/12/2014] [Indexed: 01/23/2023] Open
Abstract
Drosophila Enhancer of split M8, an effector of Notch signaling, is regulated by protein kinase CK2. The phosphatase PP2A is thought to play an opposing (inhibitory) role, but the identity of the regulatory subunit was unknown. The studies described here reveal a role for the PP2A regulatory subunit widerborst (wdb) in three developmental contexts; the bristle, wing and the R8 photoreceptors of the eye. wdb overexpression elicits bristle and wing defects akin to reduced Notch signaling, whereas hypomorphic mutations in this PP2A subunit elicit opposite effects. We have also evaluated wdb functions using mutations in Notch and E(spl) that affect the eye. We find that the eye and R8 defects of the well-known Nspl mutation are enhanced by a hypomorphic allele of wdb, whereas they are strongly rescued by wdb overexpression. Similarly, ectopic wdb rescues the eye and R8 defects of the E(spl)D mutation, which affects the m8 gene. In addition, wdb overexpression also rescues the bristle defects of ectopically expressed M8, or the eye and R8 defects of its CK2 phosphomimetic variant M8-S159D. The latter finding suggests that PP2A may target M8 at highly conserved residues in the vicinity of the CK2 site, whose phosphorylation controls repression of Atonal and the R8 fate. Together, the studies identify PP2A-Wdb as a participant in Notch signaling, and suggest that M8 activity is controlled by phosphorylation and dephosphorylation. The conservation of the phosphorylation sites between Drosophila E(spl) and the HES/HER proteins from mammals, reptiles, amphibians, birds and fish raises the prospect that this mode of regulation is widespread.
Collapse
|
32
|
Liu M, Wang C, Li D, Liu Y, Sheng Q, Lv Z, Yu W, Wang D, Zhang Y, Nie Z. Cloning and expression characteristics of the notch-associated gene BmE(spl)mγ from silkworm, Bombyx mori. Appl Biochem Biotechnol 2014; 173:2065-75. [PMID: 24916802 DOI: 10.1007/s12010-014-1003-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 05/26/2014] [Indexed: 10/25/2022]
Abstract
The E(spl)mγ gene in Drosophila is a regulatory target gene downstream of the Notch pathway. BmE(spl)mγ (Bombyx mori, E(spl)mγ) is an ortholog of the Drosophila E(spl)mγ gene, and the gene encodes a protein with 248 amino acid residues. This gene was cloned and overexpressed in Escherichia coli BL21(DE3). The recombinant protein was purified and subsequently used to generate a rabbit polyclonal antibody. Western blotting analyses showed that BmE(spl)mγ expression is high in pupa and egg, and low in larva and moth. In the fifth instar larva, the protein levels are high in head, epidermis, sexual gland, trachea, and the fatbody and low in the Malpighian tubule, hemolymph, gut, and silk gland. The further immunohistochemical analyses also showed higher BmE(spl)mγ expression in the head of fifth instar larva and pupa. Of the four moth parts studied, the thorax had the highest expression level. Thus, BmE(spl)mγ might be associated with neurogenesis in silkworm. Furthermore, DAPT (a γ-secretase inhibitor and an indirect inhibitor of Notch) blocking experiments showed that higher concentrations of the blocking agent and a longer processing time reduce the transcription levels of the BmE(spl)mγ gene, demonstrating that the silkworm BmE(spl)mγ gene is associated with the Notch signal pathway. These findings suggest that the function of BmE(spl)mγ may be similar to that of its Drosophila homolog.
Collapse
Affiliation(s)
- Min Liu
- College of Life Sciences, Zhejiang Sci-Tech University, Hanghzou, 310018, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen HF, Pan XL, Wang JW, Kong HM, Fu YM. Protein–drug interactome analysis of SSRI-mediated neurorecovery following stroke. Biosystems 2014; 120:1-9. [DOI: 10.1016/j.biosystems.2014.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/03/2014] [Accepted: 03/18/2014] [Indexed: 11/26/2022]
|
34
|
Zacharioudaki E, Bray SJ. Tools and methods for studying Notch signaling in Drosophila melanogaster. Methods 2014; 68:173-82. [PMID: 24704358 PMCID: PMC4059942 DOI: 10.1016/j.ymeth.2014.03.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/23/2014] [Accepted: 03/25/2014] [Indexed: 01/08/2023] Open
Abstract
Notch signaling involves a highly conserved pathway that mediates communication between neighboring cells. Activation of Notch by its ligands, results in the release of the Notch intracellular domain (NICD), which enters the nucleus and regulates transcription. This pathway has been implicated in many developmental decisions and diseases (including cancers) over the past decades. The simplicity of the Notch pathway in Drosophila melanogaster, in combination with the availability of powerful genetics, make this an attractive model for studying fundamental principles of Notch regulation and function. In this article we present some of the established and emerging tools that are available to monitor and manipulate the Notch pathway in Drosophila and discuss their strengths and weaknesses.
Collapse
Affiliation(s)
- Evanthia Zacharioudaki
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Sarah J Bray
- Department of Physiology Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK.
| |
Collapse
|
35
|
Andrade-Zapata I, Baonza A. The bHLH factors extramacrochaetae and daughterless control cell cycle in Drosophila imaginal discs through the transcriptional regulation of the Cdc25 phosphatase string. PLoS Genet 2014; 10:e1004233. [PMID: 24651265 PMCID: PMC3961188 DOI: 10.1371/journal.pgen.1004233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 01/27/2014] [Indexed: 11/18/2022] Open
Abstract
One of the major issues in developmental biology is about having a better understanding of the mechanisms that regulate organ growth. Identifying these mechanisms is essential to understand the development processes that occur both in physiological and pathological conditions, such as cancer. The E protein family of basic helix-loop helix (bHLH) transcription factors, and their inhibitors the Id proteins, regulate cell proliferation in metazoans. This notion is further supported because the activity of these factors is frequently deregulated in cancerous cells. The E protein orthologue Daughterless (Da) and the Id orthologue Extramacrochaetae (Emc) are the only members of these classes of bHLH proteins in Drosophila. Although these factors are involved in controlling proliferation, the mechanism underlying this regulatory activity is poorly understood. Through a genetic analysis, we show that during the development of epithelial cells in the imaginal discs, the G2/M transition, and hence cell proliferation, is controlled by Emc via Da. In eukaryotic cells, the main activator of this transition is the Cdc25 phosphatase, string. Our genetic analyses reveal that the ectopic expression of string in cells with reduced levels of Emc or high levels of Da is sufficient to rescue the proliferative defects seen in these mutant cells. Moreover, we present evidence demonstrating a role of Da as a transcriptional repressor of string. Taken together, these findings define a mechanism through which Emc controls cell proliferation by regulating the activity of Da, which transcriptionally represses string. Precise control of cell proliferation is critical for normal development and tissue homeostasis. Members of the inhibitor of differentiation (Id) family of helix-loop-helix (HLH) proteins are key regulators that coordinate the balance between cell division and differentiation. These proteins exert this function in part by combining with ubiquitously expressed bHLH transcription factors (E proteins), preventing these transcription factors from forming functional hetero- or homodimeric DNA binding complexes. Deregulation of the activity of Id proteins frequently leads to tumour formation. The Daughterless (Da) and Extramacrochaetae (Emc) proteins are the only members of the E and Id families in Drosophila, yet their role in the control of cell proliferation has not been determined. In this study, we show that the elimination of emc or the ectopic expression of da arrests cells in the G2 phase of the cell cycle. Moreover, we demonstrate that emc controls cell proliferation via Da, which acts as a transcriptional repressor of the Cdc25 phosphatase string. These results provide an important insight into the mechanisms through which Id and E protein interactions control cell cycle progression and therefore how the disruption of the function of Id proteins can induce oncogenic transformation.
Collapse
Affiliation(s)
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM), Madrid, Spain
- * E-mail:
| |
Collapse
|
36
|
Eroglu E, Burkard TR, Jiang Y, Saini N, Homem CCF, Reichert H, Knoblich JA. SWI/SNF complex prevents lineage reversion and induces temporal patterning in neural stem cells. Cell 2014; 156:1259-1273. [PMID: 24630726 DOI: 10.1016/j.cell.2014.01.053] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 11/11/2013] [Accepted: 01/16/2014] [Indexed: 12/22/2022]
Abstract
Members of the SWI/SNF chromatin-remodeling complex are among the most frequently mutated genes in human cancer, but how they suppress tumorigenesis is currently unclear. Here, we use Drosophila neuroblasts to demonstrate that the SWI/SNF component Osa (ARID1) prevents tumorigenesis by ensuring correct lineage progression in stem cell lineages. We show that Osa induces a transcriptional program in the transit-amplifying population that initiates temporal patterning, limits self-renewal, and prevents dedifferentiation. We identify the Prdm protein Hamlet as a key component of this program. Hamlet is directly induced by Osa and regulates the progression of progenitors through distinct transcriptional states to limit the number of transit-amplifying divisions. Our data provide a mechanistic explanation for the widespread tumor suppressor activity of SWI/SNF. Because the Hamlet homologs Evi1 and Prdm16 are frequently mutated in cancer, this mechanism could well be conserved in human stem cell lineages. PAPERCLIP:
Collapse
Affiliation(s)
- Elif Eroglu
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Thomas R Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Yanrui Jiang
- Biozentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Nidhi Saini
- Biozentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Catarina C F Homem
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Heinrich Reichert
- Biozentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|
37
|
|
38
|
Fu AQ, Russell S, Bray SJ, Tavaré S. Bayesian clustering of replicated time-course gene expression data with weak signals. Ann Appl Stat 2013. [DOI: 10.1214/13-aoas650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
39
|
Giagtzoglou N, Li T, Yamamoto S, Bellen HJ. Drosophila EHBP1 regulates Scabrous secretion during Notch-mediated lateral inhibition. J Cell Sci 2013; 126:3686-96. [PMID: 23788431 DOI: 10.1242/jcs.126292] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Notch signaling is an evolutionarily conserved pathway that plays a central role in numerous developmental and disease processes. The versatility of the Notch pathway relies on the activity of context-dependent regulators. These include rab11, sec15, arp3 and Drosophila EHBP1 (dEHBP1), which control Notch signaling and cell fate acquisition in asymmetrically dividing mechanosensory lineages by regulating the trafficking of the ligand Delta. Here, we show that dEHBP1 also controls the specification of R8 photoreceptors, as its loss results in the emergence of supernumerary R8 photoreceptors. Given the requirements for Notch signaling during lateral inhibition, we propose that dEHBP1 regulates distinct aspects of Notch signaling in different developmental contexts. We show that dEHBP1 regulates the exocytosis of Scabrous, a positive regulator of Notch signaling. In conclusion, dEHBP1 provides developmental versatility of intercellular signaling by regulating the trafficking of distinct Notch signaling components.
Collapse
Affiliation(s)
- Nikolaos Giagtzoglou
- Department of Molecular and Human Genetics, Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
40
|
Kux K, Kiparaki M, Delidakis C. The two Tribolium E(spl) genes show evolutionarily conserved expression and function during embryonic neurogenesis. Mech Dev 2013; 130:207-25. [PMID: 23485410 DOI: 10.1016/j.mod.2013.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 01/05/2023]
Abstract
Tribolium castaneum is a well-characterised model insect, whose short germ-band mode of embryonic development is characteristic of many insect species and differs from the exhaustively studied Drosophila. Mechanisms of early neurogenesis, however, show significant conservation with Drosophila, as a characteristic pattern of neuroblasts arises from neuroectoderm proneural clusters in response to the bHLH activator Ash, a homologue of Achaete-Scute. Here we study the expression and function of two other bHLH proteins, the bHLH-O repressors E(spl)1 and E(spl)3. Their Drosophila homologues are expressed in response to Notch signalling and antagonize the activity of Achaete-Scute proteins, thus restricting the number of nascent neuroblasts. E(spl)1 and 3 are the only E(spl) homologues in Tribolium and both show expression in the cephalic and ventral neuroectoderm during embryonic neurogenesis, as well as a dynamic pattern of expression in other tissues. Their expression starts early, soon after Ash expression and is dependent on both Ash and Notch activities. They act redundantly, since a double E(spl) knockdown (but not single knockdowns) results in neurogenesis defects similar to those caused by Notch loss-of-function. A number of other activities have been evolutionarily conserved, most notably their ability to interact with proneural proteins Scute and Daughterless.
Collapse
Affiliation(s)
- Kristina Kux
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas and Department of Biology, University of Crete, Heraklion, Crete, Greece
| | | | | |
Collapse
|
41
|
van de Hoef DL, Bonner JM, Boulianne GL. FKBP14 is an essential gene that regulates Presenilin protein levels and Notch signaling in Drosophila. Development 2013; 140:810-9. [PMID: 23318643 DOI: 10.1242/dev.081356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Presenilins were identified as causative factors in familial Alzheimer's disease and also play an essential role in Notch signaling during development. We previously identified FKBP14, a member of the family of FK506-binding proteins (FKBPs), as a modifier of Presenilin in Drosophila. FKBPs are highly conserved peptidyl-prolyl cis-trans isomerases that play integral roles in protein folding, assembly and trafficking. Although FKBPs have been implicated in a broad range of biological processes, they are non-essential in yeast and their role in the development of multicellular organisms remains unclear. We show that FKBP14 is an essential gene in Drosophila and that loss of FKBP14 gives rise to specific defects in eye, bristle and wing development. FKBP14 mutants genetically interact with components of the Notch pathway, indicating that these phenotypes are associated, at least in part, with dysregulation of Notch signaling. We show that whereas Notch trafficking to the membrane is unaffected in FKBP14 mutants, levels of Notch target genes are reduced, suggesting that FKBP14 acts downstream of Notch activation at the membrane. Consistent with this model, we find that Presenilin protein levels and γ-secretase activity are reduced in FKBP14 null mutants. Altogether, our data demonstrate that FKBP14 plays an essential role in development, one aspect of which includes regulating members of the Notch signaling pathway.
Collapse
Affiliation(s)
- Diana L van de Hoef
- The Hospital for Sick Children, Program in Developmental and Stem Cell Biology and Department of Molecular Genetics, University of Toronto, MaRS Toronto Medical Discovery Tower, 101 College Street, Room 12-305, Toronto, ON M5G 1L7, Canada.
| | | | | |
Collapse
|
42
|
Pérez-Gómez R, Slováková J, Rives-Quinto N, Krejci A, Carmena A. A Serrate-Notch-Canoe complex mediates glial-neuroepithelial cell interactions essential during Drosophila optic lobe development. J Cell Sci 2013; 126:4873-84. [DOI: 10.1242/jcs.125617] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
It is firmly established that neuron-glia interactions are fundamental across species for the correct establishment of a functional brain. Here, we found that the glia of the Drosophila larval brain display an essential non-autonomous role during the development of the optic lobe. The optic lobe develops from neuroepithelial cells that proliferate by dividing symmetrically until they switch to asymmetric/differentiative divisions generating neuroblasts. The proneural gene lethal of scute (l'sc) is transiently activated by the Epidermal Growth Factor Receptor (EGFR)/Ras signal transduction pathway at the leading edge of a proneural wave that sweeps from medial to lateral neuroepithelium promoting this switch. This process is tightly regulated by the tissue-autonomous function within the neuroepithelium of multiple signaling pathways, including EGFR/Ras and Notch. This study shows that the Notch ligand Serrate (Ser) is expressed in the glia and it forms a complex in vivo with Notch and Canoe, which colocalize at the adherens junctions of neuroepithelial cells. This complex is crucial for glial-neuroepithelial cell interactions during optic lobe development. Ser is tissue-autonomously required in the glia where it activates Notch to regulate its proliferation, and non-autonomously in the neuroepithelium where Ser induces Notch signaling to avoid the premature activation of the EGFR/Ras pathway and hence of L'sc. Interestingly, different Notch activity reporters showed very different expression patterns in the glia and in the neuroepithelium, suggesting the existence of tissue-specific factors that promote the expression of particular Notch target genes or/and a reporter response dependent on different thresholds of Notch signaling.
Collapse
|
43
|
Choi YH, Ann EJ, Yoon JH, Mo JS, Kim MY, Park HS. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) enhances osteoclast differentiation via the up-regulation of Notch1 protein stability. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:69-79. [DOI: 10.1016/j.bbamcr.2012.10.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 10/11/2012] [Accepted: 10/16/2012] [Indexed: 11/29/2022]
|
44
|
Abstract
Notch is a receptor that mediates cell-cell interactions in animal development, and aberrations in Notch signal transduction can cause cancer and other human diseases. Here, I describe the major advances in the Notch field from the identification of the first mutant in Drosophila almost a century ago through the elucidation of the unusual mechanism of signal transduction a little over a decade ago. As an essay for the GENETICS Perspectives series, it is my personal and critical commentary as well as an historical account of discovery.
Collapse
|
45
|
Abstract
An intricate interplay of signalling pathways dictates the acquisition of specific cell fates during development. The NOTCH receptor is the central element in a cell-interaction mechanism that controls the fate of a very broad spectrum of precursor cells. Conservation across species implies that signalling through this receptor is a tool frequently used by metazoans to modulate the fate of precursor cells. This article describes recent advances in the genetic and molecular dissection of this developmentally fundamental pathway that have provided new insights into the mechanism by which extracellular signals act through the NOTCH receptor to determine or alter cellular fate.
Collapse
Affiliation(s)
- R J Fleming
- Dept of Biology, The University of Rochester, Hutchinson Hall - River Campus, Rochester, NY 14627, USA
| | | | | |
Collapse
|
46
|
Ann EJ, Kim HY, Seo MS, Mo JS, Kim MY, Yoon JH, Ahn JS, Park HS. Wnt5a controls Notch1 signaling through CaMKII-mediated degradation of the SMRT corepressor protein. J Biol Chem 2012; 287:36814-29. [PMID: 22888005 DOI: 10.1074/jbc.m112.356048] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serine-threonine Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is the key component in noncanonical Wnt5a signaling and has been shown to regulate its signaling. In this study, we found that CaMKII induced by Wnt5a remarkably reduced the protein stability of the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT), a co-repressor of Notch signaling, through proteasomal degradation. Wnt5a was found to enhance Notch1 intracellular domain (Notch1-IC) transcription activity, which could be inhibited by treatment with KN93, a CaMKII inhibitor. The kinase activity of CaMKII was essential for the activation of Notch signaling. We also determined that CaMKII could enhance the association between Notch1-IC and RBP-Jk. Furthermore, the physical association between RBP-Jk and SMRT was substantially suppressed by CaMKII. We demonstrated that CaMKII directly bound and phosphorylated SMRT at Ser-1407, thereby facilitating SMRT translocation from the nucleus to the cytoplasm and proteasome-dependent degradation. These results suggest that CaMKII down-regulated the protein stability of SMRT through proteasomal degradation.
Collapse
Affiliation(s)
- Eun-Jung Ann
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
San Juan BP, Andrade-Zapata I, Baonza A. The bHLH factors Dpn and members of the E(spl) complex mediate the function of Notch signalling regulating cell proliferation during wing disc development. Biol Open 2012; 1:667-76. [PMID: 23213460 PMCID: PMC3507296 DOI: 10.1242/bio.20121172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Notch signalling pathway plays an essential role in the intricate control of cell proliferation and pattern formation in many organs during animal development. In addition, mutations in most members of this pathway are well characterized and frequently lead to tumour formation. The Drosophila imaginal wing discs have provided a suitable model system for the genetic and molecular analysis of the different pathway functions. During disc development, Notch signalling at the presumptive wing margin is necessary for the restricted activation of genes required for pattern formation control and disc proliferation. Interestingly, in different cellular contexts within the wing disc, Notch can either promote cell proliferation or can block the G1-S transition by negatively regulating the expression of dmyc and bantam micro RNA. The target genes of Notch signalling that are required for these functions have not been identified. Here, we show that the Hes vertebrate homolog, deadpan (dpn), and the Enhancer-of-split complex (E(spl)C) genes act redundantly and cooperatively to mediate the Notch signalling function regulating cell proliferation during wing disc development.
Collapse
Affiliation(s)
- Beatriz P San Juan
- Centro de Biología Molecular Severo Ochoa (CSIC/UAM) C/Nicolás Cabrera 1 , 28049 Madrid , Spain
| | | | | |
Collapse
|
48
|
The SUMO pathway promotes basic helix-loop-helix proneural factor activity via a direct effect on the Zn finger protein senseless. Mol Cell Biol 2012; 32:2849-60. [PMID: 22586269 DOI: 10.1128/mcb.06595-11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During development, proneural transcription factors of the basic helix-loop-helix (bHLH) family are required to commit cells to a neural fate. In Drosophila neurogenesis, a key mechanism promoting sense organ precursor (SOP) fate is the synergy between proneural factors and their coactivator Senseless in transcriptional activation of target genes. Here we present evidence that posttranslational modification by SUMO enhances this synergy via an effect on Senseless protein. We show that Senseless is a direct target for SUMO modification and that mutagenesis of a predicted SUMOylation motif in Senseless reduces Senseless/proneural synergy both in vivo and in cell culture. We propose that SUMOylation of Senseless via lysine 509 promotes its synergy with proneural proteins during transcriptional activation and hence regulates an important step in neurogenesis leading to the formation and maturation of the SOPs.
Collapse
|
49
|
Zacharioudaki E, Magadi SS, Delidakis C. bHLH-O proteins are crucial for Drosophila neuroblast self-renewal and mediate Notch-induced overproliferation. Development 2012; 139:1258-69. [PMID: 22357926 DOI: 10.1242/dev.071779] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Drosophila larval neurogenesis is an excellent system for studying the balance between self-renewal and differentiation of a somatic stem cell (neuroblast). Neuroblasts (NBs) give rise to differentiated neurons and glia via intermediate precursors called GMCs or INPs. We show that E(spl)mγ, E(spl)mβ, E(spl)m8 and Deadpan (Dpn), members of the basic helix-loop-helix-Orange protein family, are expressed in NBs but not in differentiated cells. Double mutation for the E(spl) complex and dpn severely affects the ability of NBs to self-renew, causing premature termination of proliferation. Single mutations produce only minor defects, which points to functional redundancy between E(spl) proteins and Dpn. Expression of E(spl)mγ and m8, but not of dpn, depends on Notch signalling from the GMC/INP daughter to the NB. When Notch is abnormally activated in NB progeny cells, overproliferation defects are seen. We show that this depends on the abnormal induction of E(spl) genes. In fact E(spl) overexpression can partly mimic Notch-induced overproliferation. Therefore, E(spl) and Dpn act together to maintain the NB in a self-renewing state, a process in which they are assisted by Notch, which sustains expression of the E(spl) subset.
Collapse
Affiliation(s)
- Evanthia Zacharioudaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Department of Biology, University of Crete, Heraklion, Crete, Greece
| | | | | |
Collapse
|
50
|
Kim SM, Kim MY, Ann EJ, Mo JS, Yoon JH, Park HS. Presenilin-2 regulates the degradation of RBP-Jk protein through p38 mitogen-activated protein kinase. J Cell Sci 2012; 125:1296-308. [PMID: 22302987 DOI: 10.1242/jcs.095984] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Transcriptional regulation performs a central role in Notch1 signaling by recombining binding protein Suppressor of Hairless (RBP-Jk)--a signaling pathway that is widely involved in determination of cell fate. Our earlier work demonstrated the possible regulation of the Notch1-RBP-Jk pathway through protein degradation of RBP-Jk; however, the potential regulator for the degradation of RBP-Jk remains to be determined. Here, we report that the expression of endogenous and exogenous RBP-Jk was increased significantly in cells treated with proteasome- and lysosome-specific inhibitors. The effects of these inhibitors on RBP-Jk occurred in a dose- and time-dependent manner. The level of RBP-Jk protein was higher in presenilin-2 (PS2)-knockout cells than in presenilin-1 (PS1)-knockout cells. Furthermore, the level of RBP-Jk was decreased by expression of PS2 in PS1 and PS2 double-knockout cells. We also found that PS1-knockout cells treated with a specific inhibitor of p38 mitogen-activated protein kinase ∂ (MAPK) had significantly increased levels of RBP-Jk. p38 MAPK phosphorylates RBP-Jk at Thr339 by physical binding, which subsequently induces the degradation and ubiquitylation of the RBP-Jk protein. Collectively, our results indicate that PS2 modulates the degradation of RBP-Jk through phosphorylation by p38 MAPK.
Collapse
Affiliation(s)
- Su-Man Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, 500-757, Republic of Korea
| | | | | | | | | | | |
Collapse
|