1
|
Kroll KL, Sosnick TR, Rock RS. Design and Use of AsLOV2-Based Optogenetic Tools for Actin Imaging. Methods Mol Biol 2025; 2840:89-100. [PMID: 39724346 DOI: 10.1007/978-1-0716-4047-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
We present protocols for using an optogenetic tool called LILAC for actin imaging. LILAC is a light-controlled version of Lifeact that uses the Avena sativa LOV2 (AsLOV2) domain. By significantly reducing Lifeact's affinity for the cytoskeleton in the dark, LILAC reduces concentration-dependent negative side effects while enabling new image processing methods. We discuss the considerations for using this probe of live-cell actin dynamics, including fluorescent protein selection, cell maintenance, microscopy protocols, and image processing. Our work highlights the potential of AsLOV2-based optogenetics for novel imaging and control tools in cell biology.
Collapse
Affiliation(s)
- Kourtney L Kroll
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
| | - Tobin R Sosnick
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology & The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. J Cell Biol 2024; 223:e202406119. [PMID: 39373700 PMCID: PMC11461286 DOI: 10.1083/jcb.202406119] [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: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Two protocadherins, Dachsous and Fat, regulate organ growth in Drosophila via the Hippo pathway. Dachsous and Fat bind heterotypically to regulate the abundance and subcellular localization of a "core complex" consisting of Dachs, Dlish, and Approximated. This complex localizes to the junctional cortex where it represses Warts. Dachsous is believed to promote growth by recruiting and stabilizing this complex, while Fat represses growth by promoting its degradation. Here, we examine the functional relationships between the intracellular domains of Dachsous and Fat and the core complex. While Dachsous promotes the accumulation of core complex proteins in puncta, it is not required for their assembly. Indeed, the core complex accumulates maximally in the absence of both Dachsous and Fat. Furthermore, Dachsous represses growth in the absence of Fat by removing the core complex from the junctional cortex. Fat similarly recruits core complex components but promotes their degradation. Our findings reveal that Dachsous and Fat coordinately constrain tissue growth by repressing the core complex.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
3
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599638. [PMID: 38948705 PMCID: PMC11212998 DOI: 10.1101/2024.06.18.599638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Two protocadherins, Dachsous (Ds) and Fat (Ft), regulate organ growth in Drosophila via the Hippo pathway. Ds and Ft bind heterotypically to regulate the abundance and subcellular localization of a 'core complex' consisting of Dachs, Dlish and Approximated. This complex localizes to the junctional cortex where it promotes growth by repressing the pathway kinase Warts. Ds is believed to promote growth by recruiting and stabilizing the core complex at the junctional cortex, while Ft represses growth by promoting degradation of core complex components. Here, we examine the functions of intracellular domains of Ds and Ft and their relationship to the core complex. While Ds promotes accumulation of the core complex proteins in cortical puncta, it is not required for core complex assembly. Indeed, the core complex assembles maximally in the absence of both Ds and Ft. Furthermore, while Ds promotes growth in the presence of Ft, it represses growth in the absence of Ft by removing the core complex from the junctional cortex. Ft similarly recruits core complex components, however it normally promotes their degradation. Our findings reveal that Ds and Ft constrain tissue growth by repressing the default 'on' state of the core complex.
Collapse
|
4
|
Nishio S, Emori C, Wiseman B, Fahrenkamp D, Dioguardi E, Zamora-Caballero S, Bokhove M, Han L, Stsiapanava A, Algarra B, Lu Y, Kodani M, Bainbridge RE, Komondor KM, Carlson AE, Landreh M, de Sanctis D, Yasumasu S, Ikawa M, Jovine L. ZP2 cleavage blocks polyspermy by modulating the architecture of the egg coat. Cell 2024; 187:1440-1459.e24. [PMID: 38490181 PMCID: PMC10976854 DOI: 10.1016/j.cell.2024.02.013] [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: 05/29/2023] [Revised: 11/07/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
Following the fertilization of an egg by a single sperm, the egg coat or zona pellucida (ZP) hardens and polyspermy is irreversibly blocked. These events are associated with the cleavage of the N-terminal region (NTR) of glycoprotein ZP2, a major subunit of ZP filaments. ZP2 processing is thought to inactivate sperm binding to the ZP, but its molecular consequences and connection with ZP hardening are unknown. Biochemical and structural studies show that cleavage of ZP2 triggers its oligomerization. Moreover, the structure of a native vertebrate egg coat filament, combined with AlphaFold predictions of human ZP polymers, reveals that two protofilaments consisting of type I (ZP3) and type II (ZP1/ZP2/ZP4) components interlock into a left-handed double helix from which the NTRs of type II subunits protrude. Together, these data suggest that oligomerization of cleaved ZP2 NTRs extensively cross-links ZP filaments, rigidifying the egg coat and making it physically impenetrable to sperm.
Collapse
Affiliation(s)
- Shunsuke Nishio
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Chihiro Emori
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Benjamin Wiseman
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Dirk Fahrenkamp
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Elisa Dioguardi
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Marcel Bokhove
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Ling Han
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Alena Stsiapanava
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Blanca Algarra
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Yonggang Lu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Mayo Kodani
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Rachel E Bainbridge
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kayla M Komondor
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anne E Carlson
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Cell and Molecular Biology, Uppsala University, 75124 Uppsala, Sweden
| | | | - Shigeki Yasumasu
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Tokyo, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| | - Luca Jovine
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
5
|
Lobb-Rabe M, Nawrocka WI, Zhang R, Ashley J, Carrillo RA, Özkan E. Neuronal Wiring Receptors Dprs and DIPs Are GPI Anchored and This Modification Contributes to Their Cell Surface Organization. eNeuro 2024; 11:ENEURO.0184-23.2023. [PMID: 38233143 PMCID: PMC10863630 DOI: 10.1523/eneuro.0184-23.2023] [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: 05/26/2023] [Revised: 11/20/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024] Open
Abstract
The Drosophila Dpr and DIP proteins belong to the immunoglobulin superfamily of cell surface proteins (CSPs). Their hetero- and homophilic interactions have been implicated in a variety of neuronal functions, including synaptic connectivity, cell survival, and axon fasciculation. However, the signaling pathways underlying these diverse functions are unknown. To gain insight into Dpr-DIP signaling, we sought to examine how these CSPs are associated with the membrane. Specifically, we asked whether Dprs and DIPs are integral membrane proteins or membrane anchored through the addition of glycosylphosphatidylinositol (GPI) linkage. We demonstrate that most Dprs and DIPs are GPI anchored to the membrane of insect cells and validate these findings for some family members in vivo using Drosophila larvae, where GPI anchor cleavage results in loss of surface labeling. Additionally, we show that GPI cleavage abrogates aggregation of insect cells expressing cognate Dpr-DIP partners. To test if the GPI anchor affects Dpr and DIP localization, we replaced it with a transmembrane domain and observed perturbation of subcellular localization on motor neurons and muscles. These data suggest that membrane anchoring of Dprs and DIPs through GPI linkage is required for localization and that Dpr-DIP intracellular signaling likely requires transmembrane coreceptors.
Collapse
Affiliation(s)
- Meike Lobb-Rabe
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637
- Program in Cell and Molecular Biology, The University of Chicago, Chicago, Illinois 60637
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
| | - Wioletta I Nawrocka
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637
| | - Ruiling Zhang
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, Illinois 60637
| | - James Ashley
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
| | - Robert A Carrillo
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois 60637
- Program in Cell and Molecular Biology, The University of Chicago, Chicago, Illinois 60637
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
| | - Engin Özkan
- Neuroscience Institute, The University of Chicago, Chicago, Illinois 60637
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois 60637
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
6
|
Tokamov SA, Buiter S, Ullyot A, Scepanovic G, Williams AM, Fernandez-Gonzalez R, Horne-Badovinac S, Fehon RG. Cortical tension promotes Kibra degradation via Par-1. Mol Biol Cell 2024; 35:ar2. [PMID: 37903240 PMCID: PMC10881160 DOI: 10.1091/mbc.e23-06-0246] [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: 06/26/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/01/2023] Open
Abstract
The Hippo pathway is an evolutionarily conserved regulator of tissue growth. Multiple Hippo signaling components are regulated via proteolytic degradation. However, how these degradation mechanisms are themselves modulated remains unexplored. Kibra is a key upstream pathway activator that promotes its own ubiquitin-mediated degradation upon assembling a Hippo signaling complex. Here, we demonstrate that Hippo complex-dependent Kibra degradation is modulated by cortical tension. Using classical genetic, osmotic, and pharmacological manipulations of myosin activity and cortical tension, we show that increasing cortical tension leads to Kibra degradation, whereas decreasing cortical tension increases Kibra abundance. Our study also implicates Par-1 in regulating Kib abundance downstream of cortical tension. We demonstrate that Par-1 promotes ubiquitin-mediated Kib degradation in a Hippo complex-dependent manner and is required for tension-induced Kib degradation. Collectively, our results reveal a previously unknown molecular mechanism by which cortical tension affects Hippo signaling and provide novel insights into the role of mechanical forces in growth control.
Collapse
Affiliation(s)
- Sherzod A. Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Stephan Buiter
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Anne Ullyot
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Audrey Miller Williams
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Rodrigo Fernandez-Gonzalez
- Institute of Biomedical Engineering and Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5G 1M1, Canada
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
7
|
Tokamov SA, Nouri N, Rich A, Buiter S, Glotzer M, Fehon RG. Apical polarity and actomyosin dynamics control Kibra subcellular localization and function in Drosophila Hippo signaling. Dev Cell 2023; 58:1864-1879.e4. [PMID: 37729921 PMCID: PMC10591919 DOI: 10.1016/j.devcel.2023.08.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/02/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
The Hippo pathway is an evolutionarily conserved regulator of tissue growth that integrates inputs from both polarity and actomyosin networks. An upstream activator of the Hippo pathway, Kibra, localizes at the junctional and medial regions of the apical cortex in epithelial cells, and medial accumulation promotes Kibra activity. Here, we demonstrate that cortical Kibra distribution is controlled by a tug-of-war between apical polarity and actomyosin dynamics. We show that while the apical polarity network, in part via atypical protein kinase C (aPKC), tethers Kibra at the junctional cortex to silence its activity, medial actomyosin flows promote Kibra-mediated Hippo complex formation at the medial cortex, thereby activating the Hippo pathway. This study provides a mechanistic understanding of the relationship between the Hippo pathway, polarity, and actomyosin cytoskeleton, and it offers novel insights into how fundamental features of epithelial tissue architecture can serve as inputs into signaling cascades that control tissue growth, patterning, and morphogenesis.
Collapse
Affiliation(s)
- Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Nicki Nouri
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Ashley Rich
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Stephan Buiter
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
8
|
Kroll KL, French AR, Sosnick TR, Rock RS. LILAC: enhanced actin imaging with an optogenetic Lifeact. Nat Methods 2023; 20:214-217. [PMID: 36717692 PMCID: PMC10986358 DOI: 10.1038/s41592-022-01761-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
Lifeact is a popular peptide-based label of actin filaments in live cells. We have designed an improved Lifeact variant, LILAC, that binds to actin in light using the LOV2 protein. Light control allows the user to modulate actin labeling, enabling image analysis that leverages modulation for an enhanced view of F-actin dynamics in cells. Furthermore, the tool reduces actin perturbations and cell sickness caused by Lifeact overexpression.
Collapse
Affiliation(s)
- Kourtney L Kroll
- Biophysical Sciences Graduate Program, The University of Chicago, Chicago, IL, USA
| | - Alexander R French
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Tobin R Sosnick
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA.
| | - Ronald S Rock
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Yoo TJ, Sup Shim M, Bang J, Kim JH, Jae Lee B. SPS1 deficiency-triggered PGRP-LC and Toll expression controls innate immunity in Drosophila S2 cells. Biol Open 2022; 11:275744. [PMID: 35723425 PMCID: PMC9364239 DOI: 10.1242/bio.059295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/14/2022] [Indexed: 12/29/2022] Open
Abstract
Selenophosphate synthetase 1 (SPS1) is an essential gene for the cell growth and embryogenesis in Drosophila melanogaster. We have previously reported that SPS1 deficiency stimulates the expression of genes responsible for the innate immune system, including antimicrobial peptides (AMPs), in Drosophila S2 cells. However, the underlying mechanism has not been elucidated. Here, we investigated the immune pathways that control the SPS1-deficiency-induced expression of AMPs in S2 cells. It was found that the activation of AMP expression is regulated by both immune deficiency (IMD) and the Toll pathway. Double knockdown of the upstream genes of each pathway with SPS1 showed that the peptidoglycan recognition protein-LC (PGRP-LC) and Toll genes are targeted by SPS1 for regulating these pathways. We also found that the IMD and Toll pathway regulate AMP expression by cross-talking. The levels of PGRP-LC and Toll mRNAs were upregulated upon Sps1 knockdown (6.4±0.36 and 3.2±0.45-fold, respectively, n=3). Overexpression of each protein also upregulated AMPs. Interestingly, PGRP-LC overexpression upregulated AMP more than Toll overexpression. These data strongly suggest that SPS1 controls the innate immune system of D. melanogaster through regulating PGRP-LC and Toll expression.
Collapse
Affiliation(s)
- Tack-Jin Yoo
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Myoung Sup Shim
- Department of Ophthalmology, Duke Eye Center, Duke Eye Center, Duke University, Durham, NC 27705, USA
| | - Jeyoung Bang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Byeong Jae Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea,Author for correspondence ()
| |
Collapse
|
10
|
18F Site-Specific Labelling of a Single-Chain Antibody against Activated Platelets for the Detection of Acute Thrombosis in Positron Emission Tomography. Int J Mol Sci 2022; 23:ijms23136886. [PMID: 35805892 PMCID: PMC9267009 DOI: 10.3390/ijms23136886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
Positron emission tomography is the imaging modality of choice when it comes to the high sensitivity detection of key markers of thrombosis and inflammation, such as activated platelets. We, previously, generated a fluorine-18 labelled single-chain antibody (scFv) against ligand-induced binding sites (LIBS) on activated platelets, binding it to the highly abundant platelet glycoprotein integrin receptor IIb/IIIa. We used a non-site-specific bio conjugation approach with N-succinimidyl-4-[18F]fluorobenzoate (S[18F]FB), leading to a mixture of products with reduced antigen binding. In the present study, we have developed and characterised a novel fluorine-18 PET radiotracer, based on this antibody, using site-specific bio conjugation to engineer cysteine residues with N-[2-(4-[18F]fluorobenzamido)ethyl]maleimide ([18F]FBEM). ScFvanti-LIBS and control antibody mut-scFv, with engineered C-terminal cysteine, were reduced, and then, they reacted with N-[2-(4-[18F]fluorobenzamido)ethyl]maleimide ([18F]FBEM). Radiolabelled scFv was injected into mice with FeCl3-induced thrombus in the left carotid artery. Clots were imaged in a PET MR imaging system, and the amount of radioactivity in major organs was measured using an ionisation chamber and image analysis. Assessment of vessel injury, as well as the biodistribution of the radiolabelled scFv, was studied. In the in vivo experiments, we found uptake of the targeted tracer in the injured vessel, compared with the non-injured vessel, as well as a high uptake of both tracers in the kidney, lung, and muscle. As expected, both tracers cleared rapidly via the kidney. Surprisingly, a large quantity of both tracers was taken up by organs with a high glutathione content, such as the muscle and lung, due to the instability of the maleimide cysteine bond in vivo, which warrants further investigations. This limits the ability of the novel antibody radiotracer 18F-scFvanti-LIBS to bind to the target in vivo and, therefore, as a useful agent for the sensitive detection of activated platelets. We describe the first fluorine-18 variant of the scFvanti-LIBS against activated platelets using site-specific bio conjugation.
Collapse
|
11
|
Zheng Q, Gao N, Sun Q, Li X, Wang Y, Xiao H. bfc, a novel serpent co-factor for the expression of croquemort, regulates efferocytosis in Drosophila melanogaster. PLoS Genet 2021; 17:e1009947. [PMID: 34860835 PMCID: PMC8673676 DOI: 10.1371/journal.pgen.1009947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 12/15/2021] [Accepted: 11/15/2021] [Indexed: 01/01/2023] Open
Abstract
Efferocytosis is the process by which phagocytes recognize, engulf, and digest (or clear) apoptotic cells during development. Impaired efferocytosis is associated with developmental defects and autoimmune diseases. In Drosophila melanogaster, recognition of apoptotic cells requires phagocyte surface receptors, including the scavenger receptor CD36-related protein, Croquemort (Crq, encoded by crq). In fact, Crq expression is upregulated in the presence of apoptotic cells, as well as in response to excessive apoptosis. Here, we identified a novel gene bfc (booster for croquemort), which plays a role in efferocytosis, specifically the regulation of the crq expression. We found that Bfc protein interacts with the zinc finger domain of the GATA transcription factor Serpent (Srp), to enhance its direct binding to the crq promoter; thus, they function together in regulating crq expression and efferocytosis. Overall, we show that Bfc serves as a Srp co-factor to upregulate the transcription of the crq encoded receptor, and consequently boosts macrophage efferocytosis in response to excessive apoptosis. Therefore, this study clarifies how phagocytes integrate apoptotic cell signals to mediate efferocytosis.
Collapse
Affiliation(s)
- Qian Zheng
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Ning Gao
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Qiling Sun
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Xiaowen Li
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Yanzhe Wang
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Hui Xiao
- College of Life Sciences, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
12
|
To V, Kim HJ, Jang W, Sreejith P, Kim C. Lin28 and Imp are Required for Stability of Bowl Transcripts in Hub
Cells of the Drosophila Testis. Dev Reprod 2021; 25:313-319. [PMID: 35141457 PMCID: PMC8807131 DOI: 10.12717/dr.2021.25.4.313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/28/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022]
Abstract
Hub cells comprise a niche for germline stem cells and cyst stem cells in the
Drosophila testis. Hub cells arise from common somatic
gonadal precursors in embryos, but the mechanism of their specification is still
poorly understood. Here we find that RNA binding proteins Lin28 and Imp mediate
transcript stability of Bowl, a known hub specification factor; Bowl transcripts
were reduced in the testis of Lin28 and Imp mutants, and also when RNA-mediated
interference against Lin28 or Imp was expressed in hub cells. In tissue culture
Luciferase assays involving the Bowl 3’UTR, stability of Luc reporter
transcripts depended on the Bowl 3’UTR and required Lin28 and Imp. Our
findings suggest that proper Bowl function during hub cell specification
requires Lin28 and Imp in the testis hub cells.
Collapse
Affiliation(s)
- Van To
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Hyun Ju Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Wijeong Jang
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | | | - Changsoo Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
- Corresponding author Changsoo Kim, School of
Biological Sciences and Technology, Chonnam National University, Gwangju 61186,
Korea. Tel: +82-62-530-5201, E-mail:
| |
Collapse
|
13
|
Malik S. Negative regulation of diminutive cancer regulator through differentiation and microRNA pathway components in Drosophila cells. ACTA ACUST UNITED AC 2021; 45:180-186. [PMID: 33907499 PMCID: PMC8068763 DOI: 10.3906/biy-2012-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/26/2021] [Indexed: 11/03/2022]
Abstract
Drosophila model is intensively studied for the development of cancer. The diminutive (dMyc), a homolog of the human MYC gene, is responsible for cell- apoptosis and its upregulation is responsible for determining the fate of cancerous growth in humans and Drosophila model. This work implores the requirement of dMyc and its expression as one of the major regulator of cancer with other proteins and repression of dMyc mRNA in Drosophila S2 cells. Here we report protein complex of Argonaute 1 (AGO1), Bag of marbles (Bam), and Brain tumor (Brat) proteins and not the individual factor of this complex repression of dMyc mRNA in Drosophila Schneider 2 cells and promote differentiation in cystoblast of Drosophila ovary. These results exhibit the significant role of this complex, including master differentiation factor Bam with other various differentiation factor Brat and microRNA pathway component AGO1, which may negatively regulate dMyc mRNA and so the dMyc protein.
Collapse
Affiliation(s)
- Sumira Malik
- Amity institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand India.,School of Biological Sciences and Technology, Chonnam National University the Republic of Korea
| |
Collapse
|
14
|
Tokamov SA, Su T, Ullyot A, Fehon RG. Negative feedback couples Hippo pathway activation with Kibra degradation independent of Yorkie-mediated transcription. eLife 2021; 10:62326. [PMID: 33555257 PMCID: PMC7895526 DOI: 10.7554/elife.62326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
The Hippo (Hpo) pathway regulates tissue growth in many animals. Multiple upstream components promote Hpo pathway activity, but the organization of these different inputs, the degree of crosstalk between them, and whether they are regulated in a distinct manner is not well understood. Kibra (Kib) activates the Hpo pathway by recruiting the core Hpo kinase cassette to the apical cortex. Here, we show that the Hpo pathway downregulates Drosophila Kib levels independently of Yorkie-mediated transcription. We find that Hpo signaling complex formation promotes Kib degradation via SCFSlimb-mediated ubiquitination, that this effect requires Merlin, Salvador, Hpo, and Warts, and that this mechanism functions independently of other upstream Hpo pathway activators. Moreover, Kib degradation appears patterned by differences in mechanical tension across the wing. We propose that Kib degradation mediated by Hpo pathway components and regulated by cytoskeletal tension serves to control Kib-driven Hpo pathway activation and ensure optimally scaled and patterned tissue growth.
Collapse
Affiliation(s)
- Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States.,Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, United States
| | - Ting Su
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Anne Ullyot
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, United States.,Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, United States
| |
Collapse
|
15
|
Casler JC, Zajac AL, Valbuena FM, Sparvoli D, Jeyifous O, Turkewitz AP, Horne-Badovinac S, Green WN, Glick BS. ESCargo: a regulatable fluorescent secretory cargo for diverse model organisms. Mol Biol Cell 2020; 31:2892-2903. [PMID: 33112725 PMCID: PMC7927198 DOI: 10.1091/mbc.e20-09-0591] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/24/2022] Open
Abstract
Membrane traffic can be studied by imaging a cargo protein as it transits the secretory pathway. The best tools for this purpose initially block export of the secretory cargo from the endoplasmic reticulum (ER) and then release the block to generate a cargo wave. However, previously developed regulatable secretory cargoes are often tricky to use or specific for a single model organism. To overcome these hurdles for budding yeast, we recently optimized an artificial fluorescent secretory protein that exits the ER with the aid of the Erv29 cargo receptor, which is homologous to mammalian Surf4. The fluorescent secretory protein forms aggregates in the ER lumen and can be rapidly disaggregated by addition of a ligand to generate a nearly synchronized cargo wave. Here we term this regulatable secretory protein ESCargo (Erv29/Surf4-dependent secretory cargo) and demonstrate its utility not only in yeast cells, but also in cultured mammalian cells, Drosophila cells, and the ciliate Tetrahymena thermophila. Kinetic studies indicate that rapid export from the ER requires recognition by Erv29/Surf4. By choosing an appropriate ER signal sequence and expression vector, this simple technology can likely be used with many model organisms.
Collapse
Affiliation(s)
- Jason C. Casler
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Allison L. Zajac
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Fernando M. Valbuena
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Daniela Sparvoli
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Okunola Jeyifous
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Aaron P. Turkewitz
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - William N. Green
- Department of Neurobiology, University of Chicago, Chicago, IL 60637
- Marine Biological Laboratory, Woods Hole, MA 02543
| | - Benjamin S. Glick
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
16
|
Rich A, Fehon RG, Glotzer M. Rho1 activation recapitulates early gastrulation events in the ventral, but not dorsal, epithelium of Drosophila embryos. eLife 2020; 9:56893. [PMID: 33200987 PMCID: PMC7717907 DOI: 10.7554/elife.56893] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/16/2020] [Indexed: 12/27/2022] Open
Abstract
Ventral furrow formation, the first step in Drosophila gastrulation, is a well-studied example of tissue morphogenesis. Rho1 is highly active in a subset of ventral cells and is required for this morphogenetic event. However, it is unclear whether spatially patterned Rho1 activity alone is sufficient to recapitulate all aspects of this morphogenetic event, including anisotropic apical constriction and coordinated cell movements. Here, using an optogenetic probe that rapidly and robustly activates Rho1 in Drosophila tissues, we show that Rho1 activity induces ectopic deformations in the dorsal and ventral epithelia of Drosophila embryos. These perturbations reveal substantial differences in how ventral and dorsal cells, both within and outside the zone of Rho1 activation, respond to spatially and temporally identical patterns of Rho1 activation. Our results demonstrate that an asymmetric zone of Rho1 activity is not sufficient to recapitulate ventral furrow formation and reveal that additional, ventral-specific factors contribute to the cell- and tissue-level behaviors that emerge during ventral furrow formation.
Collapse
Affiliation(s)
- Ashley Rich
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, United States
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, United States
| | - Michael Glotzer
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, United States
| |
Collapse
|
17
|
Malik S, Jang W, Kim JY, Kim C. Mechanisms ensuring robust repression of the Drosophila female germline stem cell maintenance factor Nanos via posttranscriptional regulation. FASEB J 2020; 34:11421-11430. [PMID: 32654316 DOI: 10.1096/fj.202000656r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/22/2022]
Abstract
During oogenesis in the Drosophila ovary, numerous translational regulators promote the self-renewal or differentiation of stem cells. An intriguing question is how these regulators combine to execute translational regulation. Here, we study mechanisms for the posttranscriptional regulation of nos, a critical stem cell self-renewal factor in the Drosophila ovary; specifically, regulators that promote differentiation of the stem cell daughter. Previous studies showed that Bam, Bgcn, Mei-P26, and Sxl form a complex and repress nos expression through the nos 3'UTR. To further elucidate mechanistic processes of Nos translational regulation, we reconstituted nos repression in cultured Drosophila cells. We identify Ago1 and Brat as new members, and show that Ago1 acts through miRNA binding sites in the proximal region of the nos 3'UTR, whereas Sxl acts via an Sxl binding sequence in the distal region. Combining these findings with published reports, we propose that additional factors Bam, Bgcn, Mei-P26, and Brat are recruited to nos mRNAs through interaction with Ago1 and Sxl. These findings elucidate mechanisms of nos regulation by diverse translational repressors.
Collapse
Affiliation(s)
- Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea
| | - Ji Young Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
18
|
Site-Specific Glycation and Chemo-enzymatic Antibody Sortagging for the Retargeting of rAAV6 to Inflamed Endothelium. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:261-269. [PMID: 31453264 PMCID: PMC6704353 DOI: 10.1016/j.omtm.2019.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
Gene therapy holds great potential for conditions such as cardiovascular disease, including atherosclerosis and also vascular cancers, yet available vectors such as the adeno-associated virus (rAAV) transduce the vasculature poorly. To enable retargeting, a single-chain antibody (scFv) that binds to the vascular cell-adhesion molecule (VCAM-1) overexpressed at areas of endothelial inflammation was site specifically and covalently conjugated to the exterior of rAAV6. To achieve conjugation, the scFv was functionalized with an orthogonal click chemistry group. This conjugation utilized site-specific sortase A methodology, thus preserving scFv binding capacity to VCAM-1. The AAV6 was separately functionalized with 4-azidophenyl glyoxal (APGO) via covalent adducts to arginine residues in the capsid’s heparin co-receptor binding region. APGO functionalization removed native tropism, greatly reducing rAAV6-GFP transduction into all cells tested, and the effect was similar to the inhibition seen in the presence of heparin. Utilizing the incorporated functionalizations, the scFv was then covalently conjugated to the exterior of rAAV6 via strain-promoted azide-alkyne cycloaddition (SPAAC). With both the removal of native heparin tropism and the addition of VCAM-1 targeting, rAAV6 transduction of endothelial cells was greatly enhanced compared to control cells. Thus, this novel and modular targeting system could have further application in re-directing AAV6 toward inflamed endothelium for therapeutic use.
Collapse
|
19
|
The target specificity of the RNA binding protein Pumilio is determined by distinct co-factors. Biosci Rep 2019; 39:BSR20190099. [PMID: 31097674 PMCID: PMC6549094 DOI: 10.1042/bsr20190099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/17/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022] Open
Abstract
Puf family proteins are translational regulators essential to a wide range of biological processes, including cell fate specification, stem cell self-renewal, and neural function. Yet, despite being associated with hundreds of RNAs, the underlying mechanisms of Puf target specification remain to be fully elucidated. In Drosophila, Pumilio – a sole Puf family protein – is known to collaborate with cofactors Nanos (Nos) and Brain Tumor (Brat); however, their roles in target specification are not clearly defined. Here, we identify Bag-of-marbles (Bam) as a new Pum cofactor in repression of Mothers against dpp (mad) mRNAs, for which Nos is known to be dispensable. Notably, our data show that Nos (but not Bam) was required for Pum association with hunchback (hb) mRNAs, a well-known target of Pum and Nos. In contrast, Bam (but not Nos) was required for Pum association with mad mRNAs. These findings show for the first time that Pum target specificity is determined not independently but in collaboration with cofactors.
Collapse
|
20
|
Wu W, Zhai M, Li C, Yu X, Song X, Gao S, Li B. Multiple functions of miR-8-3p in the development and metamorphosis of the red flour beetle, Tribolium castaneum. INSECT MOLECULAR BIOLOGY 2019; 28:208-221. [PMID: 30230097 DOI: 10.1111/imb.12539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The microRNA miR-8-3p is conserved among insects and closely involved in development and immunity, but its functions in vivo are unexplored in the red flour beetle, Tribolium castaneum. Here, we show that miR-8-3p was highly expressed in late larva and early adult stages, as determined by quantitative real-time PCR. It was enriched in the fat body and cuticle in late larval tissues and abundant in the head and cuticle in early adult tissues, indicating this microRNA plays important roles during T. castaneum development. Specific inhibition of miR-8-3p in late larvae led to metamorphosis defects in the development of wings, eyes, legs and embryo. Moreover, a series of genes related to organism development were identified as miR-8-3p targets by computational prediction and microRNA-messenger RNA interaction validation, including Wingless, Eyg, Fpps and Sema-1a. These genes were critical for the regulation of the larva-to-adult transition. Eyg, as a functional target of miR-8-3p, participates in eye development, which was further confirmed by luciferase assay and loss-of-function analyses. In brief, miR-8-3p is broadly involved in the development of wings, eyes and legs through its target genes and has extensive regulatory roles during T. castaneum development.
Collapse
Affiliation(s)
- W Wu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - M Zhai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - C Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - X Yu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - X Song
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - S Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - B Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
21
|
Okamoto N, Viswanatha R, Bittar R, Li Z, Haga-Yamanaka S, Perrimon N, Yamanaka N. A Membrane Transporter Is Required for Steroid Hormone Uptake in Drosophila. Dev Cell 2018; 47:294-305.e7. [PMID: 30293839 PMCID: PMC6219898 DOI: 10.1016/j.devcel.2018.09.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/06/2018] [Accepted: 09/09/2018] [Indexed: 02/08/2023]
Abstract
Steroid hormones are a group of lipophilic hormones that are believed to enter cells by simple diffusion to regulate diverse physiological processes through intracellular nuclear receptors. Here, we challenge this model in Drosophila by demonstrating that Ecdysone Importer (EcI), a membrane transporter identified from two independent genetic screens, is involved in cellular uptake of the steroid hormone ecdysone. EcI encodes an organic anion transporting polypeptide of the evolutionarily conserved solute carrier organic anion superfamily. In vivo, EcI loss of function causes phenotypes indistinguishable from ecdysone- or ecdysone receptor (EcR)-deficient animals, and EcI knockdown inhibits cellular uptake of ecdysone. Furthermore, EcI regulates ecdysone signaling in a cell-autonomous manner and is both necessary and sufficient for inducing ecdysone-dependent gene expression in culture cells expressing EcR. Altogether, our results challenge the simple diffusion model for cellular uptake of ecdysone and may have wide implications for basic and medical aspects of steroid hormone studies.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Raghuvir Viswanatha
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Riyan Bittar
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Zhongchi Li
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Sachiko Haga-Yamanaka
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
22
|
Xu J, Vanderzalm PJ, Ludwig M, Su T, Tokamov SA, Fehon RG. Yorkie Functions at the Cell Cortex to Promote Myosin Activation in a Non-transcriptional Manner. Dev Cell 2018; 46:271-284.e5. [PMID: 30032991 PMCID: PMC6086586 DOI: 10.1016/j.devcel.2018.06.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/09/2018] [Accepted: 06/19/2018] [Indexed: 02/06/2023]
Abstract
The Hippo signaling pathway is an evolutionarily conserved mechanism that controls organ size in animals. Yorkie is well known as a transcriptional co-activator that functions downstream of the Hippo pathway to positively regulate transcription of genes that promote tissue growth. Recent studies have shown that increased myosin activity activates both Yorkie and its vertebrate orthologue YAP, resulting in increased nuclear localization and tissue growth. Here we show that Yorkie also can accumulate at the cell cortex in the apical junctional region. Moreover, Yorkie functions at the cortex to promote activation of myosin through a myosin regulatory light chain kinase, Stretchin-Mlck. This Yorkie function is not dependent on its transcriptional activity and is required for larval and adult tissues to achieve appropriate size. Based on these results, we suggest that Yorkie functions in a feedforward "amplifier" loop that promotes myosin activation, and thereby greater Yorkie activity, in response to tension.
Collapse
Affiliation(s)
- Jiajie Xu
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Pamela J Vanderzalm
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Biology, John Carroll University, University Heights, OH 44118, USA
| | - Michael Ludwig
- Department of Ecology and Evolutionary Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Ting Su
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Sherzod A Tokamov
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA; Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
23
|
Heigwer F, Port F, Boutros M. RNA Interference (RNAi) Screening in Drosophila. Genetics 2018; 208:853-874. [PMID: 29487145 PMCID: PMC5844339 DOI: 10.1534/genetics.117.300077] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/28/2017] [Indexed: 12/22/2022] Open
Abstract
In the last decade, RNA interference (RNAi), a cellular mechanism that uses RNA-guided degradation of messenger RNA transcripts, has had an important impact on identifying and characterizing gene function. First discovered in Caenorhabditis elegans, RNAi can be used to silence the expression of genes through introduction of exogenous double-stranded RNA into cells. In Drosophila, RNAi has been applied in cultured cells or in vivo to perturb the function of single genes or to systematically probe gene function on a genome-wide scale. In this review, we will describe the use of RNAi to study gene function in Drosophila with a particular focus on high-throughput screening methods applied in cultured cells. We will discuss available reagent libraries and cell lines, methodological approaches for cell-based assays, and computational methods for the analysis of high-throughput screens. Furthermore, we will review the generation and use of genome-scale RNAi libraries for tissue-specific knockdown analysis in vivo and discuss the differences and similarities with the use of genome-engineering methods such as CRISPR/Cas9 for functional analysis.
Collapse
Affiliation(s)
- Florian Heigwer
- Division of Signaling and Functional Genomics, German Cancer Research Center, and Department of Cell and Molecular Biology, Heidelberg University, Medical Faculty Mannheim, D-69120, Germany
| | - Fillip Port
- Division of Signaling and Functional Genomics, German Cancer Research Center, and Department of Cell and Molecular Biology, Heidelberg University, Medical Faculty Mannheim, D-69120, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center, and Department of Cell and Molecular Biology, Heidelberg University, Medical Faculty Mannheim, D-69120, Germany
| |
Collapse
|
24
|
Malik S, Jang W, Kim C. Protein Interaction Mapping of Translational Regulators Affecting Expression of the Critical Stem Cell Factor Nos. Dev Reprod 2017; 21:449-456. [PMID: 29354790 PMCID: PMC5769139 DOI: 10.12717/dr.2017.21.4.449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 12/26/2017] [Accepted: 12/27/2017] [Indexed: 01/01/2023]
Abstract
The germline stem cells of the Drosophila ovary continuously
produce eggs throughout the life- span. Intricate regulation of stemness and
differentiation is critical to this continuous production. The translational
regulator Nos is an intrinsic factor that is required for maintenance of
stemness in germline stem cells. Nos expression is reduced in differentiating
cells at the post-transcriptional level by diverse translational regulators.
However, molecular mechanisms underlying Nos repression are not completely
understood. Through three distinct protein-protein interaction experiments, we
identified specific molecular interactions between translational regulators
involved in Nos repression. Our findings suggest a model in which protein
complexes assemble on the 3’ untranslated region of Nos mRNA in order to
regulate Nos expression at the post-transcriptional level.
Collapse
Affiliation(s)
- Sumira Malik
- Hormone Research Center, School of Biological Sciences and Technology, College of Natural Sciences,Chonnam National University, Gwangju 61186, Republic of Korea.,Dept. of Agriculture and Forestry, Tula's Institute Dhoolkot, Dehradun-248011, India
| | - Wijeong Jang
- Hormone Research Center, School of Biological Sciences and Technology, College of Natural Sciences,Chonnam National University, Gwangju 61186, Republic of Korea
| | - Changsoo Kim
- Hormone Research Center, School of Biological Sciences and Technology, College of Natural Sciences,Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
25
|
Pimmett VL, Deng H, Haskins JA, Mercier RJ, LaPointe P, Simmonds AJ. The activity of the Drosophila Vestigial protein is modified by Scalloped-dependent phosphorylation. Dev Biol 2017; 425:58-69. [PMID: 28322734 DOI: 10.1016/j.ydbio.2017.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 02/01/2017] [Accepted: 03/14/2017] [Indexed: 12/18/2022]
Abstract
The Drosophila vestigial gene is required for proliferation and differentiation of the adult wing and for differentiation of larval and adult muscle identity. Vestigial is part of a multi-protein transcription factor complex, which includes Scalloped, a TEAD-class DNA binding protein. Binding Scalloped is necessary for translocation of Vestigial into the nucleus. We show that Vestigial is extensively post-translationally modified and at least one of these modifications is required for proper function during development. We have shown that there is p38-dependent phosphorylation of Serine 215 in the carboxyl-terminal region of Vestigial. Phosphorylation of Serine 215 occurs in the nucleus and requires the presence of Scalloped. Comparison of a phosphomimetic and non-phosphorylatable mutant forms of Vestigial shows differences in the ability to rescue the wing and muscle phenotypes associated with a null vestigial allele.
Collapse
Affiliation(s)
- Virginia L Pimmett
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7
| | - Hua Deng
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7; Howard Hughes Medical Institute, Dept. of Physiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Julie A Haskins
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7
| | - Rebecca J Mercier
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7
| | - Paul LaPointe
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7
| | - Andrew J Simmonds
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada T6G2H7
| |
Collapse
|
26
|
Matakatsu H, Blair SS, Fehon RG. The palmitoyltransferase Approximated promotes growth via the Hippo pathway by palmitoylation of Fat. J Cell Biol 2016; 216:265-277. [PMID: 28031421 PMCID: PMC5223609 DOI: 10.1083/jcb.201609094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/19/2016] [Accepted: 12/09/2016] [Indexed: 01/07/2023] Open
Abstract
The palmitoyl transferase Approximated regulates signaling by the protocadherin Fat to control tissue growth upstream of the Hippo pathway in Drosophila. Matakatsu et al. show that palmitoylation of the intracellular domain of Fat by Approximated negatively regulates Fat and its ability to restrict growth. The large protocadherin Fat functions to promote Hippo pathway activity in restricting tissue growth. Loss of Fat leads to accumulation of the atypical myosin Dachs at the apical junctional region, which in turn promotes growth by inhibiting Warts. We previously identified Approximated (App), a DHHC domain palmitoyltransferase, as a negative regulator of Fat signaling in growth control. We show here that App promotes growth by palmitoylating the intracellular domain of Fat, and that palmitoylation negatively regulates Fat function. Independently, App also recruits Dachs to the apical junctional region through protein–protein association, thereby stimulating Dachs’s activity in promoting growth. Further, we show that palmitoylation by App functions antagonistically to phosphorylation by Discs-overgrown, which activates Fat. Together, these findings suggest a model in which App promotes Dachs activity by simultaneously repressing Fat via posttranslational modification and recruiting Dachs to the apical junctional region, thereby promoting tissue growth.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637.,Department of Zoology, University of Wisconsin, Madison, WI 53706
| | - Seth S Blair
- Department of Zoology, University of Wisconsin, Madison, WI 53706
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
27
|
Rao T, Gao R, Takada S, Al Abo M, Chen X, Walters KJ, Pommier Y, Aihara H. Novel TDP2-ubiquitin interactions and their importance for the repair of topoisomerase II-mediated DNA damage. Nucleic Acids Res 2016; 44:10201-10215. [PMID: 27543075 PMCID: PMC5137425 DOI: 10.1093/nar/gkw719] [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] [Received: 01/20/2016] [Revised: 07/30/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022] Open
Abstract
Tyrosyl DNA phosphodiesterase 2 (TDP2) is a multifunctional protein implicated in DNA repair, signal transduction and transcriptional regulation. In its DNA repair role, TDP2 safeguards genome integrity by hydrolyzing 5′-tyrosyl DNA adducts formed by abortive topoisomerase II (Top2) cleavage complexes to allow error-free repair of DNA double-strand breaks, thereby conferring cellular resistance against Top2 poisons. TDP2 consists of a C-terminal catalytic domain responsible for its phosphodiesterase activity, and a functionally uncharacterized N-terminal region. Here, we demonstrate that this N-terminal region contains a ubiquitin (Ub)-associated (UBA) domain capable of binding multiple forms of Ub with distinct modes of interactions and preference for either K48- or K63-linked polyUbs over monoUb. The structure of TDP2 UBA bound to monoUb shows a canonical mode of UBA-Ub interaction. However, the absence of the highly conserved MGF motif and the presence of a fourth α-helix make TDP2 UBA distinct from other known UBAs. Mutations in the TDP2 UBA-Ub binding interface do not affect nuclear import of TDP2, but severely compromise its ability to repair Top2-mediated DNA damage, thus establishing the importance of the TDP2 UBA–Ub interaction in DNA repair. The differential binding to multiple Ub forms could be important for responding to DNA damage signals under different contexts or to support the multi-functionality of TDP2.
Collapse
Affiliation(s)
- Timsi Rao
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rui Gao
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Saeko Takada
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Muthana Al Abo
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiang Chen
- Protein Processing Section, Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Kylie J Walters
- Protein Processing Section, Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Yves Pommier
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Abstract
RNA interference (RNAi) is a potent tool for perturbation of gene function in model organisms and human cells. In Drosophila, efficient RNAi enables screening approaches for components of cellular processes in vivo and in vitro. In cultured cells, measuring the effect of depleting gene products on a genome-wide scale can systematically associate gene function with diverse processes, such as cell growth and proliferation, signaling and trafficking. Here, we describe methods for RNAi experiments in cultured Drosophila cells with a focus on genome-wide loss-of-function screening. We illustrate the design of long double-stranded RNAs and provide protocols for their production by in vitro transcription and delivery in cell-based assays. Furthermore, we provide methods to fine-tune signaling reporters and high-content microscopy assays for genome-wide screening. Finally, we describe essential steps of high-throughput data analysis and how the experimental set-up can improve data normalization using a genome-wide RNAi screen for Wnt pathway activity data as an example.
Collapse
Affiliation(s)
- Maximilian Billmann
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg University, Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | - Michael Boutros
- Division of Signaling and Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg University, Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany.
| |
Collapse
|
29
|
Delbeke EIP, Lozach O, Le Gall T, Berchel M, Montier T, Jaffrès PA, Van Geem KM, Stevens CV. Evaluation of the transfection efficacies of quaternary ammonium salts prepared from sophorolipids. Org Biomol Chem 2016; 14:3744-51. [DOI: 10.1039/c6ob00241b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Two quaternary ammonium sophorolipids proved to be suitable as transfection vectors for gene delivery.
Collapse
Affiliation(s)
- E. I. P. Delbeke
- SynBioC
- Department of Sustainable Organic Chemistry and Technology
- Ghent University
- 9000 Ghent
- Belgium
| | - O. Lozach
- Université de Brest
- CEMCA
- CNRS UMR 6521
- IBSAM
- 29238 Brest
| | - T. Le Gall
- IBiSA SynNanoVect platform
- IBSAM
- Université de Brest
- Faculté de médecine Morvan
- avenue Camille Desmoulins
| | - M. Berchel
- Université de Brest
- CEMCA
- CNRS UMR 6521
- IBSAM
- 29238 Brest
| | - T. Montier
- IBiSA SynNanoVect platform
- IBSAM
- Université de Brest
- Faculté de médecine Morvan
- avenue Camille Desmoulins
| | - P.-A. Jaffrès
- Université de Brest
- CEMCA
- CNRS UMR 6521
- IBSAM
- 29238 Brest
| | - K. M. Van Geem
- LCT
- Department of Chemical Engineering and Technical Chemistry
- Ghent University
- 9052 Ghentn
- Belgium
| | - C. V. Stevens
- SynBioC
- Department of Sustainable Organic Chemistry and Technology
- Ghent University
- 9000 Ghent
- Belgium
| |
Collapse
|
30
|
Suárez-Patiño SF, Mancini RA, Pereira CA, Suazo CAT, Mendonça RZ, Jorge SAC. Transient expression of rabies virus glycoprotein (RVGP) in Drosophila melanogaster Schneider 2 (S2) cells. J Biotechnol 2014; 192 Pt A:255-62. [DOI: 10.1016/j.jbiotec.2014.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 11/25/2022]
|
31
|
Lee GJ, Hyun S. Multiple targets of the microRNA miR-8 contribute to immune homeostasis in Drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 45:245-251. [PMID: 24694685 DOI: 10.1016/j.dci.2014.03.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 03/04/2014] [Accepted: 03/06/2014] [Indexed: 06/03/2023]
Abstract
MicroRNAs (miRNAs) are noncoding, around 22-nucleotide-long RNAs that exert significant modulatory roles in gene expression throughout the genome. As such, miRNAs take part in and modulate almost all biological processes like cell growth, development, and immunity. We previously showed that miR-8 miRNA plays a role in maintaining immune homeostasis in Drosophila. Here, we further discovered that targeting of multiple coding genes by miR-8 contributes to the maintenance of immune homeostasis. Toll and Dorsal, respectively the receptor and transcription factor in the Toll immune pathway, were found to be miR-8 targets, as shown by reporter assays and miR-8 null flies. Moreover, U-shaped (Ush), a previously verified miR-8 target, was seen to mediate miR-8 regulation of immune homeostasis. Consistently, overexpression of either Dorsal or Ush in the fat body led to increased Drosomycin expression, mimicking that induced by deletion of miR-8. Furthermore, mutation in Toll immune pathway or Ush rescues the abnormal expression of Drosomycin and lethality in miR-8 mutant. Thus, miR-8 regulates Drosophila immune homeostasis by targeting multiple immune genes, thereby contributing to survival.
Collapse
Affiliation(s)
- Gang Jun Lee
- Department of Life Science, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - Seogang Hyun
- Department of Life Science, Chung-Ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
32
|
Kim K, Yim J. Structural modelling and molecular characterization of omega-class glutathione S-transferase 2 from Drosophila melanogaster. INSECT MOLECULAR BIOLOGY 2014; 23:357-366. [PMID: 24533905 DOI: 10.1111/imb.12086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Glutathione S-transferase omega (GSTO) is a recently identified Glutathione S-transferase (GST), and it has several known functions and variable distribution patterns in many organisms. In Drosophila, GstO2 exists as two isoforms, GstO2A and GstO2B. Despite the high sequence homology between the two GstO2 isoforms, they have different physiological functions. In the present study, we characterized the structural and molecular properties of Drosophila melanogaster GstO2 isoforms. Homology modelling of GstO2s using I-TASSER servers for protein structure and function prediction revealed that the two GstO2s have different electropotential surface distributions and different shapes of the substrate-binding sites. The recombinant GstO2s have native molecular weights of ∼60 kDa. GstO2s have similar optimum conditions for enzymatic reactions at pH 8.0 and 40 °C. The kinetic parameters of the reduction of dehydroascorbate by these two GstO2s were determined. Collectively, our results provide structural insights into the different substrate profiles of the GstO2 isoforms.
Collapse
Affiliation(s)
- K Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan-si, Korea
| | | |
Collapse
|
33
|
Vigdorovich V, Ramagopal UA, Lázár-Molnár E, Sylvestre E, Lee JS, Hofmeyer KA, Zang X, Nathenson SG, Almo SC. Structure and T cell inhibition properties of B7 family member, B7-H3. Structure 2013; 21:707-17. [PMID: 23583036 DOI: 10.1016/j.str.2013.03.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/18/2013] [Accepted: 03/01/2013] [Indexed: 01/25/2023]
Abstract
T cell activity is controlled by a combination of antigen-dependent signaling through the T cell receptor and a set of auxiliary signals delivered through antigen-independent interactions, including the recognition of the B7 family of ligands. B7-H3 is a recently identified B7 family member that is strongly overexpressed in a range of cancers and correlates with poor prognosis. We report the crystal structure of murine B7-H3 at a 3 Å resolution, which provides a model for the organization of the IgV and IgC domains within the ectodomain. We demonstrate that B7-H3 inhibits T cell proliferation and show that the FG loop of the IgV domain plays a critical role in this function. B7-H3 crystallized as an unusual dimer arising from the exchange of the G strands in the IgV domains of partner molecules. This arrangement, in combination with previous reports, highlights the dynamic nature and plasticity of the immunoglobulin fold.
Collapse
Affiliation(s)
- Vladimir Vigdorovich
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The Smc5/Smc6/MAGE complex confers resistance to caffeine and genotoxic stress in Drosophila melanogaster. PLoS One 2013; 8:e59866. [PMID: 23555814 PMCID: PMC3610895 DOI: 10.1371/journal.pone.0059866] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 02/19/2013] [Indexed: 12/20/2022] Open
Abstract
The SMC5/6 protein complex consists of the Smc5, Smc6 and Non-Smc-Element (Nse) proteins and is important for genome stability in many species. To identify novel components in the DNA repair pathway, we carried out a genetic screen to identify mutations that confer reduced resistance to the genotoxic effects of caffeine, which inhibits the ATM and ATR DNA damage response proteins. This approach identified inactivating mutations in CG5524 and MAGE, homologs of genes encoding Smc6 and Nse3 in yeasts. The fact that Smc5 mutants are also caffeine-sensitive and that Mage physically interacts with Drosophila homologs of Nse proteins suggests that the structure of the Smc5/6 complex is conserved in Drosophila. Although Smc5/6 proteins are required for viability in S. cerevisiae, they are not essential under normal circumstances in Drosophila. However, flies carrying mutations in Smc5, Smc6 and MAGE are hypersensitive to genotoxic agents such as ionizing radiation, camptothecin, hydroxyurea and MMS, consistent with the Smc5/6 complex serving a conserved role in genome stability. We also show that mutant flies are not compromised for pre-mitotic cell cycle checkpoint responses. Rather, caffeine-induced apoptosis in these mutants is exacerbated by inhibition of ATM or ATR checkpoint kinases but suppressed by Rad51 depletion, suggesting a functional interaction involving homologous DNA repair pathways that deserves further scrutiny. Our insights into the SMC5/6 complex provide new challenges for understanding the role of this enigmatic chromatin factor in multi-cellular organisms.
Collapse
|
35
|
Neisch AL, Formstecher E, Fehon RG. Conundrum, an ARHGAP18 orthologue, regulates RhoA and proliferation through interactions with Moesin. Mol Biol Cell 2013; 24:1420-33. [PMID: 23468526 PMCID: PMC3639053 DOI: 10.1091/mbc.e12-11-0800] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
RhoA, a small GTPase, regulates epithelial integrity and morphogenesis by controlling filamentous actin assembly and actomyosin contractility. Another important cytoskeletal regulator, Moesin (Moe), an ezrin, radixin, and moesin (ERM) protein, has the ability to bind to and organize cortical F-actin, as well as the ability to regulate RhoA activity. ERM proteins have previously been shown to interact with both RhoGEF (guanine nucleotide exchange factors) and RhoGAP (GTPase activating proteins), proteins that control the activation state of RhoA, but the functions of these interactions remain unclear. We demonstrate that Moe interacts with an unusual RhoGAP, Conundrum (Conu), and recruits it to the cell cortex to negatively regulate RhoA activity. In addition, we show that cortically localized Conu can promote cell proliferation and that this function requires RhoGAP activity. Surprisingly, Conu's ability to promote growth also appears dependent on increased Rac activity. Our results reveal a molecular mechanism by which ERM proteins control RhoA activity and suggest a novel linkage between the small GTPases RhoA and Rac in growth control.
Collapse
Affiliation(s)
- Amanda L Neisch
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
36
|
Avanesov A, Blair SS. The Drosophila WIF1 homolog Shifted maintains glypican-independent Hedgehog signaling and interacts with the Hedgehog co-receptors Ihog and Boi. Development 2012; 140:107-16. [PMID: 23154411 DOI: 10.1242/dev.078444] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hedgehog (Hh) family proteins are secreted signaling ligands whose short- and long-range activities transform cellular fates in multiple contexts in organisms ranging from metazoans to humans. In the developing Drosophila wing, extracellular Hh binds to cell-bound glypican heparan sulfate proteoglycans (HSPGs) and the secreted protein Shifted (Shf), a member of Wnt inhibitory factor 1 (WIF1) family. The glypicans and Shf are required for long-range Hh movement and signaling; it has been proposed that Shf promotes long-range Hh signaling by reinforcing binding between Hh and the glypicans, and that much or all of glypican function in Hh signaling requires Shf. However, we will show here that Shf maintains short-range Hh signaling in the wing via a mechanism that does not require the presence of or binding to the Drosophila glypicans Dally and Dally-like protein. Conversely, we demonstrate interactions between Hh and the glypicans that are maintained, and even strengthened, in the absence of Shf. We present evidence that Shf binds to the CDO/BOC family Hh co-receptors Interference hedgehog (Ihog) and Brother of Ihog, suggesting that Shf regulates short-range Hh signaling through interactions with the receptor complex. In support of a functional interaction between Ihog and members of the Shf/WIF1 family, we show that Ihog can increase the Wnt-inhibitory activity of vertebrate WIF1; this result raises the possibility of interactions between WIF1 and vertebrate CDO/BOC family members.
Collapse
Affiliation(s)
- Andrei Avanesov
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | | |
Collapse
|
37
|
The role of glypicans in Wnt inhibitory factor-1 activity and the structural basis of Wif1's effects on Wnt and Hedgehog signaling. PLoS Genet 2012; 8:e1002503. [PMID: 22383891 PMCID: PMC3285576 DOI: 10.1371/journal.pgen.1002503] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/11/2011] [Indexed: 01/03/2023] Open
Abstract
Proper assignment of cellular fates relies on correct interpretation of Wnt and Hedgehog (Hh) signals. Members of the Wnt Inhibitory Factor-1 (WIF1) family are secreted modulators of these extracellular signaling pathways. Vertebrate WIF1 binds Wnts and inhibits their signaling, but its Drosophila melanogaster ortholog Shifted (Shf) binds Hh and extends the range of Hh activity in the developing D. melanogaster wing. Shf activity is thought to depend on reinforcing interactions between Hh and glypican HSPGs. Using zebrafish embryos and the heterologous system provided by D. melanogaster wing, we report on the contribution of glypican HSPGs to the Wnt-inhibiting activity of zebrafish Wif1 and on the protein domains responsible for the differences in Wif1 and Shf specificity. We show that Wif1 strengthens interactions between Wnt and glypicans, modulating the biphasic action of glypicans towards Wnt inhibition; conversely, glypicans and the glypican-binding “EGF-like” domains of Wif1 are required for Wif1's full Wnt-inhibiting activity. Chimeric constructs between Wif1 and Shf were used to investigate their specificities for Wnt and Hh signaling. Full Wnt inhibition required the “WIF” domain of Wif1, and the HSPG-binding EGF-like domains of either Wif1 or Shf. Full promotion of Hh signaling requires both the EGF-like domains of Shf and the WIF domains of either Wif1 or Shf. That the Wif1 WIF domain can increase the Hh promoting activity of Shf's EGF domains suggests it is capable of interacting with Hh. In fact, full-length Wif1 affected distribution and signaling of Hh in D. melanogaster, albeit weakly, suggesting a possible role for Wif1 as a modulator of vertebrate Hh signaling. In developing organisms, cells choose between alternative fates in order to make appropriately patterned tissues, and misregulation of those choices can underlie both developmental defects and cancers. Cells often make these decisions because of signals received from neighboring cells, such as those mediated by the secreted signaling proteins of the Wnt and Hedgehog (Hh) families. While signaling can be regulated by the levels of signaling or receptor proteins expressed by cells, another level of control is exerted by proteins that bind signaling proteins outside of cells and either inhibit or promote the signaling process. In the fruitfly Drosophilamelanogaster, the secreted Shifted protein has been shown to bind Hh and to increase Hh signaling, likely by reinforcing interactions between Hh and cell surface proteins of the glypican family. We provide evidence that the vertebrate homolog of Shifted, Wnt Inhibitory Factor-1 (Wif1), inhibits Wnt activity by a similar mechanism, reinforcing interactions between Wnts and glypicans in a manner that sequesters Wnts from their receptors. We also examine the structural basis for the specificities of Wif1 and Shifted for Wnt and Hh signaling, respectively, and provide evidence that Wif1, although a potent inhibitor of Wnt activity, influences D. melanogaster Hh signaling.
Collapse
|
38
|
Yang Y, Primrose DA, Leung AC, Fitzsimmons RB, McDermand MC, Missellbrook A, Haskins J, Smylie AS, Hughes SC. The PP1 phosphatase flapwing regulates the activity of Merlin and Moesin in Drosophila. Dev Biol 2011; 361:412-26. [PMID: 22133918 DOI: 10.1016/j.ydbio.2011.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 11/09/2011] [Accepted: 11/10/2011] [Indexed: 01/21/2023]
Abstract
The signalling activities of Merlin and Moesin, two closely related members of the protein 4.1 Ezrin/Radixin/Moesin family, are regulated by conformational changes. These changes are regulated in turn by phosphorylation. The same sterile 20 kinase-Slik co-regulates Merlin or Moesin activity whereby phosphorylation inactivates Merlin, but activates Moesin. Thus, the corresponding coordinate activation of Merlin and inactivation of Moesin would require coordinated phosphatase activity. We find that Drosophila melanogaster protein phosphatase type 1 β (flapwing) fulfils this role, co-regulating dephosphorylation and altered activity of both Merlin and Moesin. Merlin or Moesin are detected in a complex with Flapwing both in-vitro and in-vivo. Directed changes in flapwing expression result in altered phosphorylation of both Merlin and Moesin. These changes in the levels of Merlin and Moesin phosphorylation following reduction of flapwing expression are associated with concomitant defects in epithelial integrity and increase in apoptosis in developing tissues such as wing imaginal discs. Functionally, the defects can be partially recapitulated by over expression of proteins that mimic constitutively phosphorylated or unphosphorylated Merlin or Moesin. Our results suggest that changes in the phosphorylation levels of Merlin and Moesin lead to changes in epithelial organization.
Collapse
Affiliation(s)
- Yang Yang
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Li J, Najand N, Long W, Simmonds A. Imaging the cellular dynamics of Drosophila Argonaute proteins. Methods Mol Biol 2011; 725:143-59. [PMID: 21528452 DOI: 10.1007/978-1-61779-046-1_10] [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] [Indexed: 02/03/2023]
Abstract
Drosophila melanogaster is used extensively as a model system to uncover genetic and molecular pathways that regulate various cellular activities. There are five members of the Argonaute protein family in Drosophila. Argonautes have been found to be localized to cytoplasmic ribonucleoprotein containing structures in both cultured Drosophila cells and developing embryos. However, in fixed cell preparations some Drosophila Argonaute family proteins co-localize with structures containing known as RNA processing (P) body components while others do not. The ability to image Argonaute family proteins in live Drosophila cells, (both cultured and within developing embryos) allows for accurate genetic dissection of the pathways involved in the assembly, mobility, disassembly, and other dynamic processes of Argonaute-containing bodies. Here we describe a method of rapidly creating vectors for, and assay the activity of, fluorescently tagged Argonaute proteins in cultured Drosophila cells and embryos.
Collapse
Affiliation(s)
- Jing Li
- Faculty of Medicine and Dentistry, Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
40
|
Dicer recognizes the 5' end of RNA for efficient and accurate processing. Nature 2011; 475:201-5. [PMID: 21753850 DOI: 10.1038/nature10198] [Citation(s) in RCA: 384] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 05/16/2011] [Indexed: 11/09/2022]
Abstract
A hallmark of RNA silencing is a class of approximately 22-nucleotide RNAs that are processed from double-stranded RNA precursors by Dicer. Accurate processing by Dicer is crucial for the functionality of microRNAs (miRNAs). The current model posits that Dicer selects cleavage sites by measuring a set distance from the 3' overhang of the double-stranded RNA terminus. Here we report that human Dicer anchors not only the 3' end but also the 5' end, with the cleavage site determined mainly by the distance (∼22 nucleotides) from the 5' end (5' counting rule). This cleavage requires a 5'-terminal phosphate group. Further, we identify a novel basic motif (5' pocket) in human Dicer that recognizes the 5'-phosphorylated end. The 5' counting rule and the 5' anchoring residues are conserved in Drosophila Dicer-1, but not in Giardia Dicer. Mutations in the 5' pocket reduce processing efficiency and alter cleavage sites in vitro. Consistently, miRNA biogenesis is perturbed in vivo when Dicer-null embryonic stem cells are replenished with the 5'-pocket mutant. Thus, 5'-end recognition by Dicer is important for precise and effective biogenesis of miRNAs. Insights from this study should also afford practical benefits to the design of small hairpin RNAs.
Collapse
|
41
|
Horn T, Sandmann T, Fischer B, Axelsson E, Huber W, Boutros M. Mapping of signaling networks through synthetic genetic interaction analysis by RNAi. Nat Methods 2011; 8:341-6. [PMID: 21378980 DOI: 10.1038/nmeth.1581] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 02/04/2011] [Indexed: 12/28/2022]
Abstract
The analysis of synthetic genetic interaction networks can reveal how biological systems achieve a high level of complexity with a limited repertoire of components. Studies in yeast and bacteria have taken advantage of collections of deletion strains to construct matrices of quantitative interaction profiles and infer gene function. Yet comparable approaches in higher organisms have been difficult to implement in a robust manner. Here we report a method to identify genetic interactions in tissue culture cells through RNAi. By performing more than 70,000 pairwise perturbations of signaling factors, we identified >600 interactions affecting different quantitative phenotypes of Drosophila melanogaster cells. Computational analysis of this interaction matrix allowed us to reconstruct signaling pathways and identify a conserved regulator of Ras-MAPK signaling. Large-scale genetic interaction mapping by RNAi is a versatile, scalable approach for revealing gene function and the connectivity of cellular networks.
Collapse
Affiliation(s)
- Thomas Horn
- German Cancer Research Center (Deutsches Krebsforschungszentrum), Division Signaling and Functional Genomics and Heidelberg University, Department of Cell and Molecular Biology, Faculty of Medicine Mannheim, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Nilton A, Oshima K, Zare F, Byri S, Nannmark U, Nyberg KG, Fehon RG, Uv AE. Crooked, coiled and crimpled are three Ly6-like proteins required for proper localization of septate junction components. Development 2010; 137:2427-37. [PMID: 20570942 DOI: 10.1242/dev.052605] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular junction formation is an elaborate process that is dependent on the regulated synthesis, assembly and membrane targeting of constituting components. Here, we report on three Drosophila Ly6-like proteins essential for septate junction (SJ) formation. SJs provide a paracellular diffusion barrier and appear molecularly and structurally similar to vertebrate paranodal septate junctions. We show that Crooked (Crok), a small GPI-anchored Ly6-like protein, is required for septa formation and barrier functions. In embryos that lack Crok, SJ components are produced but fail to accumulate at the plasma membrane. Crok is detected in intracellular puncta and acts tissue-autonomously, which suggests that it resides in intracellular vesicles to assist the cell surface localization of SJ components. In addition, we demonstrate that two related Ly6 proteins, Coiled (Cold) and Crimpled (Crim), are required for SJ formation and function in a tissue-autonomous manner, and that Cold also localizes to intracellular vesicles. Specifically, Crok and Cold are required for correct membrane trafficking of Neurexin IV, a central SJ component. The non-redundant requirement for Crok, Cold, Crim and Boudin (Bou; another Ly6 protein that was recently shown to be involved in SJ formation) suggests that members of this conserved family of proteins cooperate in the assembly of SJ components, possibly by promoting core SJ complex formation in intracellular compartments associated with membrane trafficking.
Collapse
Affiliation(s)
- Anna Nilton
- Institute of Biomedicine, Göteborg University, Gothenburg, SE-40530, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Neisch AL, Speck O, Stronach B, Fehon RG. Rho1 regulates apoptosis via activation of the JNK signaling pathway at the plasma membrane. ACTA ACUST UNITED AC 2010; 189:311-23. [PMID: 20404112 PMCID: PMC2856900 DOI: 10.1083/jcb.200912010] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the absence of moesin, RhoA slips out of its normal role as a GTPase to activate the JNK MAPK pathway and spur apoptosis. Precisely controlled growth and morphogenesis of developing epithelial tissues require coordination of multiple factors, including proliferation, adhesion, cell shape, and apoptosis. RhoA, a small GTPase, is known to control epithelial morphogenesis and integrity through its ability to regulate the cytoskeleton. In this study, we examine a less well-characterized RhoA function in cell survival. We demonstrate that the Drosophila melanogaster RhoA, Rho1, promotes apoptosis independently of Rho kinase through its effects on c-Jun NH2-terminal kinase (JNK) signaling. In addition, Rho1 forms a complex with Slipper (Slpr), an upstream activator of the JNK pathway. Loss of Moesin (Moe), an upstream regulator of Rho1 activity, results in increased levels of Rho1 at the plasma membrane and cortical accumulation of Slpr. Together, these results suggest that Rho1 functions at the cell cortex to regulate JNK activity and implicate Rho1 and Moe in epithelial cell survival.
Collapse
Affiliation(s)
- Amanda L Neisch
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
44
|
Hughes SC, Formstecher E, Fehon RG. Sip1, the Drosophila orthologue of EBP50/NHERF1, functions with the sterile 20 family kinase Slik to regulate Moesin activity. J Cell Sci 2010; 123:1099-107. [PMID: 20215404 DOI: 10.1242/jcs.059469] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Organization of the plasma membrane in polarized epithelial cells is accomplished by the specific localization of transmembrane or membrane-associated proteins, which are often linked to cytoplasmic protein complexes, including the actin cytoskeleton. In this study, we identified Sip1 as a Drosophila orthologue of the ezrin-radixin-moesin (ERM) binding protein 50 (EBP50; also known as the Na(+)/H(+) exchanger regulatory factor NHERF1). In mammals, EBP50/NHERF1 is a scaffold protein required for the regulation of several transmembrane receptors and downstream signal transduction activity. In Drosophila, loss of Sip1 leads to a reduction in Slik kinase protein abundance, loss of Moesin phosphorylation and changes in epithelial structure, including mislocalization of E-cadherin and F-actin. Consistent with these findings, Moesin and Sip1 act synergistically in genetic-interaction experiments, and Sip1 protein abundance is dependent on Moesin. Co-immunoprecipitation experiments indicate that Sip1 forms a complex with both Moesin and Slik. Taken together, these data suggest that Sip1 promotes Slik-dependent phosphorylation of Moesin, and suggests a mechanism for the regulation of Moesin activity within the cell to maintain epithelial integrity.
Collapse
Affiliation(s)
- Sarah C Hughes
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.
| | | | | |
Collapse
|
45
|
Kim YI, Ryu T, Lee J, Heo YS, Ahnn J, Lee SJ, Yoo O. A genetic screen for modifiers of Drosophila caspase Dcp-1 reveals caspase involvement in autophagy and novel caspase-related genes. BMC Cell Biol 2010; 11:9. [PMID: 20100334 PMCID: PMC2822743 DOI: 10.1186/1471-2121-11-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 01/25/2010] [Indexed: 12/01/2022] Open
Abstract
Background Caspases are cysteine proteases with essential functions in the apoptotic pathway; their proteolytic activity toward various substrates is associated with the morphological changes of cells. Recent reports have described non-apoptotic functions of caspases, including autophagy. In this report, we searched for novel modifiers of the phenotype of Dcp-1 gain-of-function (GF) animals by screening promoter element- inserted Drosophila melanogaster lines (EP lines). Results We screened ~15,000 EP lines and identified 72 Dcp-1-interacting genes that were classified into 10 groups based on their functions and pathways: 4 apoptosis signaling genes, 10 autophagy genes, 5 insulin/IGF and TOR signaling pathway genes, 6 MAP kinase and JNK signaling pathway genes, 4 ecdysone signaling genes, 6 ubiquitination genes, 11 various developmental signaling genes, 12 transcription factors, 3 translation factors, and 11 other unclassified genes including 5 functionally undefined genes. Among them, insulin/IGF and TOR signaling pathway, MAP kinase and JNK signaling pathway, and ecdysone signaling are known to be involved in autophagy. Together with the identification of autophagy genes, the results of our screen suggest that autophagy counteracts Dcp-1-induced apoptosis. Consistent with this idea, we show that expression of eGFP-Atg5 rescued the eye phenotype caused by Dcp-1 GF. Paradoxically, we found that over-expression of full-length Dcp-1 induced autophagy, as Atg8b-GFP, an indicator of autophagy, was increased in the eye imaginal discs and in the S2 cell line. Taken together, these data suggest that autophagy suppresses Dcp-1-mediated apoptotic cell death, whereas Dcp-1 positively regulates autophagy, possibly through feedback regulation. Conclusions We identified a number of Dcp-1 modifiers that genetically interact with Dcp-1-induced cell death. Our results showing that Dcp-1 and autophagy-related genes influence each other will aid future investigations of the complicated relationships between apoptosis and autophagy.
Collapse
Affiliation(s)
- Young-Il Kim
- Bio Medical Research Center, Department of Biological Science, KAIST, 373-1, 305-701, Daejeon, Korea
| | | | | | | | | | | | | |
Collapse
|
46
|
Shim MS, Kim JY, Jung HK, Lee KH, Xu XM, Carlson BA, Kim KW, Kim IY, Hatfield DL, Lee BJ. Elevation of glutamine level by selenophosphate synthetase 1 knockdown induces megamitochondrial formation in Drosophila cells. J Biol Chem 2009; 284:32881-94. [PMID: 19755423 PMCID: PMC2781704 DOI: 10.1074/jbc.m109.026492] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 09/11/2009] [Indexed: 11/06/2022] Open
Abstract
Although selenophosphate synthetase 1 (SPS1/SelD) is an essential gene in Drosophila, its function has not been determined. To elucidate its intracellular role, we targeted the removal of SPS1/SelD mRNA in Drosophila SL2 cells using RNA interference technology that led to the formation of vacuole-like globular structures. Surprisingly, these structures were identified as megamitochondria, and only depolarized mitochondria developed into megamitochondria. The mRNA levels of l(2)01810 and glutamine synthetase 1 (GS1) were increased by SPS1/SelD knockdown. Blocking the expression of GS1 and l(2)01810 completely inhibited the formation of megamitochondria induced by loss of SPS1/SelD activity and decreased the intracellular levels of glutamine to those of control cells suggesting that the elevated level of glutamine is responsible for megamitochondrial formation. Overexpression of GS1 and l(2)01810 had a synergistic effect on the induction of megamitochondrial formation and on the synthesis of glutamine suggesting that l(2)01810 is involved in glutamine synthesis presumably by activating GS1. Our results indicate that, in Drosophila, SPS1/SelD regulates the intracellular glutamine by inhibiting GS1 and l(2)01810 expression and that elevated levels of glutamine lead to a nutritional stress that provides a signal for megamitochondrial formation.
Collapse
Affiliation(s)
- Myoung Sup Shim
- From the
Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Jin Young Kim
- From the
Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Hee Kyoung Jung
- From the
Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Kwang Hee Lee
- From the
Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| | - Xue-Ming Xu
- Laboratory of Cancer Prevention, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Bradley A. Carlson
- Laboratory of Cancer Prevention, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Ki Woo Kim
- National Instrumentation Center for Environmental Management, College of Agriculture and Life Science, Seoul National University, Seoul 151-742, Korea, and
| | - Ick Young Kim
- Laboratory of Cellular and Molecular Biochemistry, School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | - Dolph L. Hatfield
- Laboratory of Cancer Prevention, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Byeong Jae Lee
- From the
Laboratory of Molecular Genetics and Genomics, School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
47
|
Byun J, Yoon J, Baek K. Analysis of two promoters that control the expression of the GTP cyclohydrolase I gene in Drosophila melanogaster. Mol Cells 2009; 27:583-9. [PMID: 19387565 DOI: 10.1007/s10059-009-0072-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/03/2009] [Accepted: 03/05/2009] [Indexed: 10/20/2022] Open
Abstract
GTP cyclohydrolase I (GTPCH) is a key enzyme in the de novo synthesis of tetrahydrobiopterin. Previously, the Drosophila melanogaster GTPCH gene has been shown to be expressed from two different promoters (P1 and P2). In our study, the 5'-flanking DNA regions required for P1 and P2 promoter activities were characterized using transient expression assay. The DNA regions between -98 and +31, and between -73 and +35 are required for efficient P1 and P2 promoter activities, respectively. The regions between -98 and -56 and between -73 and -41 may contain critical elements required for the expression of GTPCH in Drosophila. By aligning the nucleotide sequences in the P1 and P2 promoter regions of the Drosophila melanogaster and Drosophila virilis GTPCH genes, several conserved elements including palindromic sequences in the regions critical for P1 and P2 promoter activities were identified. Western blot analysis of transgenic flies transformed using P1 or P2 promoter-lacZ fusion plasmids further revealed that P1 promoter expression is restricted to the late pupae and adult developmental stages but that the P2 promoter driven expression of GTPCH is constitutive throughout fly development. In addition, X-gal staining of the embryos and imaginal discs of transgenic flies suggests that the P2 promoter is active from stage 13 of embryo and is generally active in most regions of the imaginal discs at the larval stages.
Collapse
Affiliation(s)
- Jaegoo Byun
- Graduate School of Biotechnology, Kyung Hee University, 446-701 Yongin, Korea
| | | | | |
Collapse
|
48
|
Deng H, Hughes SC, Bell JB, Simmonds AJ. Alternative requirements for Vestigial, Scalloped, and Dmef2 during muscle differentiation in Drosophila melanogaster. Mol Biol Cell 2008; 20:256-69. [PMID: 18987343 DOI: 10.1091/mbc.e08-03-0288] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Vertebrate development requires the activity of the myocyte enhancer factor 2 (mef2) gene family for muscle cell specification and subsequent differentiation. Additionally, several muscle-specific functions of MEF2 family proteins require binding additional cofactors including members of the Transcription Enhancing Factor-1 (TEF-1) and Vestigial-like protein families. In Drosophila there is a single mef2 (Dmef2) gene as well single homologues of TEF-1 and vestigial-like, scalloped (sd), and vestigial (vg), respectively. To clarify the role(s) of these factors, we examined the requirements for Vg and Sd during Drosophila muscle specification. We found that both are required for muscle differentiation as loss of sd or vg leads to a reproducible loss of a subset of either cardiac or somatic muscle cells in developing embryos. This muscle requirement for Sd or Vg is cell specific, as ubiquitous overexpression of either or both of these proteins in muscle cells has a deleterious effect on muscle differentiation. Finally, using both in vitro and in vivo binding assays, we determined that Sd, Vg, and Dmef2 can interact directly. Thus, the muscle-specific phenotypes we have associated with Vg or Sd may be a consequence of alternative binding of Vg and/or Sd to Dmef2 forming alternative protein complexes that modify Dmef2 activity.
Collapse
Affiliation(s)
- Hua Deng
- Department of Cell Biology, Department of Biological Sciences, and Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | | | | | | |
Collapse
|
49
|
Mische S, He Y, Ma L, Li M, Serr M, Hays TS. Dynein light intermediate chain: an essential subunit that contributes to spindle checkpoint inactivation. Mol Biol Cell 2008; 19:4918-29. [PMID: 18799620 DOI: 10.1091/mbc.e08-05-0483] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The dynein light intermediate chain (LIC) is a subunit unique to the cytoplasmic form of dynein, but how it contributes to dynein function is not fully understood. Previous work has established that the LIC homodimer binds directly to the dynein heavy chain and may mediate the attachment of dynein to centrosomes and other cargoes. Here, we report our characterization of the LIC in Drosophila. Unlike vertebrates, in which two Lic genes encode multiple subunit isoforms, the Drosophila LIC is encoded by a single gene. We determined that the single LIC polypeptide is phosphorylated, and that different phosphoisoforms can assemble into the dynein motor complex. Our mutational analyses demonstrate that, similar to other dynein subunits, the Drosophila LIC is required for zygotic development, germline specification of the oocyte, and mitotic cell division. We show that RNA interference depletion of LIC in Drosophila S2 cells does not block the recruitment of a dynein complex to kinetochores, but it does delay inactivation of Mad2 signaling and mitotic progression. Our observations suggest the LIC contributes to a broad range of dynein functions.
Collapse
Affiliation(s)
- Sarah Mische
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | | | |
Collapse
|
50
|
Iyadurai SJP, Robinson JT, Ma L, He Y, Mische S, Li MG, Brown W, Guichard A, Bier E, Hays TS. Dynein and Star interact in EGFR signaling and ligand trafficking. J Cell Sci 2008; 121:2643-51. [PMID: 18653542 PMCID: PMC2633900 DOI: 10.1242/jcs.027144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intracellular transport and processing of ligands is critical to the activation of signal transduction pathways that guide development. Star is an essential gene in Drosophila that has been implicated in the trafficking of ligands for epidermal growth factor (EGF) receptor signaling. The role of cytoplasmic motors in the endocytic and secretory pathways is well known, but the specific requirement of motors in EGF receptor transport has not been investigated. We identified Star in a screen designed to recover second-site modifiers of the dominant rough eye phenotype of the Glued mutation Gl(1). The Glued (Gl) locus encodes the p150 subunit of the dynactin complex, an activator of cytoplasmic dynein-driven motility. We show that alleles of Gl and dynein genetically interact with both Star and EGFR alleles. Similarly to mutations in Star, the Gl(1) mutation is capable of modifying the phenotypes of the EGFR mutation Ellipse. These genetic interactions suggest a model in which Star, dynactin and dynein cooperate in the trafficking of EGF ligands. In support of this model, overexpression of the cleaved, active Spitz ligand can partially bypass defective trafficking and suppress the genetic interactions. Our direct observations of live S2 cells show that export of Spitz-GFP from the endoplasmic reticulum, as well as the trafficking of Spitz-GFP vesicles, depends on both Star and dynein.
Collapse
Affiliation(s)
- Stanley J. P. Iyadurai
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - John T. Robinson
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - Lingzhi Ma
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - Yungui He
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - Sarah Mische
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - Min-gang Li
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - William Brown
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| | - Annabel Guichard
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ethan Bier
- Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thomas S. Hays
- University of Minnesota, Department of Genetics, Cell Biology and Development, Minneapolis, MN 55455, USA
| |
Collapse
|