1
|
Arias RA, Tomlinson A. Decoding a Cell's Fate: How Notch and receptor tyrosine kinase signals specify the Drosophila R7 photoreceptor. Dev Biol 2025; 519:21-29. [PMID: 39653132 PMCID: PMC11750611 DOI: 10.1016/j.ydbio.2024.12.001] [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: 07/16/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The process by which the Drosophila R7 photoreceptor is specified has become a classic model for understanding how cell-cell signals direct cell fates. In the R7 precursor cell, both the Notch and receptor tyrosine kinase (RTK) signaling pathways are active, and the information they encode directs the specification of the R7 photoreceptor identity. In this process, Notch performs three distinct functions: it both opposes and promotes the actions of the RTK pathway to specify the photoreceptor fate, and it determines the type of photoreceptor that is specified. The RTK pathway drives transcription of phyl - a gene expression necessary for photoreceptor specification. We show that Notch activity induces transcription of the yan gene which encodes a transcriptional repressor of phyl. This defines an antagonism between the two pathways, with RTK promoting and Notch opposing phyl transcription. We previously showed that Notch activity supplies Sevenless to the R7 precursor to allow the RTK pathway hyperactivation required to overcome the Notch repression, and we now identify the regulation of Yan activity as a site of integration of RTK and Notch signaling pathways. Once the cell is specified as a photoreceptor, the third Notch function then prevents seven-up (svp) transcription. The Svp transcription factor directs the R1/6 photoreceptor fate, and the prevention of its expression ensures the default R7 specification.
Collapse
Affiliation(s)
- Ronald A Arias
- Zuckerman Institute, Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, Jerome L. Greene Science Center, Level 9 Room 028, 3227 Broadway, New York, NY, 10027, USA
| | - Andrew Tomlinson
- Zuckerman Institute, Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, Jerome L. Greene Science Center, Level 9 Room 028, 3227 Broadway, New York, NY, 10027, USA.
| |
Collapse
|
2
|
Ronald AA, Yannis EM, Tomlinson A. Roles Played by Enhancer of Split Transcription Factors in Drosophila R7 Photoreceptor Specification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635716. [PMID: 39975285 PMCID: PMC11838290 DOI: 10.1101/2025.01.30.635716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
When a cell receives multiple developmental signals simultaneously, the intracellular transduction pathways triggered by those signals are coincidentally active. How then, do the cells decode the information contained within those multiple active pathways to derive a precise developmental directive? The specification of the Drosophila R7 photoreceptor is classic model system for investigating such questions. The R7 fate is specified by the combined actions of the of the Notch (N) and receptor tyrosine kinase (RTK) signaling pathways. The two pathways cross-communicate in an integrative mechanism but also supply information independently of each other. Collectively, this information is summed to provide an unambiguous directive for the R7 fate. Our goal is to understand these mechanisms. Here, we examine how N activity represses transcription of the phyllopod gene in the process of information integration with the RTK pathway, and how it represses expression of the seven-up gene in an independent mechanism needed for R7 fate. We describe how N activity achieves these transcriptional repressions, and identify Enhancer of Split transcription factors as the mediators of those actions.
Collapse
Affiliation(s)
- A Arias Ronald
- Zuckerman Institute, Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, Jerome L. Greene Science Center, Level 9 Room 028 3227 Broadway New York, NY 10027
| | - E Mavromatakis Yannis
- Zuckerman Institute, Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, Jerome L. Greene Science Center, Level 9 Room 028 3227 Broadway New York, NY 10027
| | - Andrew Tomlinson
- Zuckerman Institute, Department of Genetics and Development, College of Physicians and Surgeons of Columbia University, Jerome L. Greene Science Center, Level 9 Room 028 3227 Broadway New York, NY 10027
| |
Collapse
|
3
|
Ma J, Wang Y, Tang X, Zhao D, Zhang D, Li C, Li W, Li T, Jiang L. TaSINA2B, interacting with TaSINA1D, positively regulates drought tolerance and root growth in wheat (Triticum aestivum L.). PLANT, CELL & ENVIRONMENT 2023; 46:3760-3774. [PMID: 37642386 DOI: 10.1111/pce.14708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/05/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023]
Abstract
Wheat (Triticum aestivum L.) is an important food crop mainly grown in arid and semiarid regions worldwide, whose productivity is severely limited by drought stress. Although various E3 ubiquitin (Ub) ligases regulate drought stress, only a few SINA-type E3 Ub ligases are known to participate in such responses. Herein, we identified and cloned 15 TaSINAs from wheat. The transcription level of TaSINA2B was highly induced by drought, osmotic and abscisic acid treatments. Two-type promoters of TaSINA2B were found in 192 wheat accessions; furthermore wheat accessions with promoter TaSINA2BII showed a considerably higher level of drought tolerance and gene expression levels than those characterizing accessions with promoter TaSINA2BI that was mainly caused by a 64 bp insertion in its promoter. Enhanced drought tolerance of TaSINA2B-overexpressing (OE) transgenic wheat lines was found to be associated with root growth promotion. Further, an interaction between TaSINA2B and TaSINA1D was detected through yeast two-hybrid and bimolecular fluorescence complementation assays. And TaSINA1D-OE transgenic wheat lines showed similar drought tolerance and root growth phenotype to those observed when TaSINA2B was overexpressed. Therefore, the variation of TaSINA2B promoter contributed to the drought stress regulation, while TaSINA2B, interacting with TaSINA1D, positively regulated drought tolerance by promoting root growth.
Collapse
Affiliation(s)
- Jianhui Ma
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Yudie Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Xiaoxiao Tang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Dongyang Zhao
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Daijing Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Chunxi Li
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Wei Li
- State Key Laboratory of Cotton Biology, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang, Henan, China
| | - Tian Li
- Key Laboratory of Crop Gene Resources and Germplasm Enhancement, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lina Jiang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
4
|
Huang Y, Yu G, Yang Y. MIGGRI: A multi-instance graph neural network model for inferring gene regulatory networks for Drosophila from spatial expression images. PLoS Comput Biol 2023; 19:e1011623. [PMID: 37939200 PMCID: PMC10659162 DOI: 10.1371/journal.pcbi.1011623] [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: 04/02/2023] [Revised: 11/20/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023] Open
Abstract
Recent breakthrough in spatial transcriptomics has brought great opportunities for exploring gene regulatory networks (GRNs) from a brand-new perspective. Especially, the local expression patterns and spatio-temporal regulation mechanisms captured by spatial expression images allow more delicate delineation of the interplay between transcript factors and their target genes. However, the complexity and size of spatial image collections pose significant challenges to GRN inference using image-based methods. Extracting regulatory information from expression images is difficult due to the lack of supervision and the multi-instance nature of the problem, where a gene often corresponds to multiple images captured from different views. While graph models, particularly graph neural networks, have emerged as a promising method for leveraging underlying structure information from known GRNs, incorporating expression images into graphs is not straightforward. To address these challenges, we propose a two-stage approach, MIGGRI, for capturing comprehensive regulatory patterns from image collections for each gene and known interactions. Our approach involves a multi-instance graph neural network (GNN) model for GRN inference, which first extracts gene regulatory features from spatial expression images via contrastive learning, and then feeds them to a multi-instance GNN for semi-supervised learning. We apply our approach to a large set of Drosophila embryonic spatial gene expression images. MIGGRI achieves outstanding performance in the inference of GRNs for early eye development and mesoderm development of Drosophila, and shows robustness in the scenarios of missing image information. Additionally, we perform interpretable analysis on image reconstruction and functional subgraphs that may reveal potential pathways or coordinate regulations. By leveraging the power of graph neural networks and the information contained in spatial expression images, our approach has the potential to advance our understanding of gene regulation in complex biological systems.
Collapse
Affiliation(s)
- Yuyang Huang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| | - Gufeng Yu
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| | - Yang Yang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| |
Collapse
|
5
|
Yang J, Mao T, Geng Z, Xue W, Ma L, Jin Y, Guo P, Qiu Z, Wang L, Yu C, Sheng Y, Zhang J, Zhang H. Constitutive expression of AtSINA2 from Arabidopsis improves grain yield, seed oil and drought tolerance in transgenic soybean. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 196:444-453. [PMID: 36758291 DOI: 10.1016/j.plaphy.2023.01.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
The SEVEN IN Absentia (SINA), a typical member of the RING E3 ligase family, plays a crucial role in plant growth, development and response to abiotic stress. However, its biological functions in oil crops are still unknown. Previously, we reported that overexpression of AtSINA2 in Arabidopsis positively regulated the drought tolerance of transgenic plants. In this work, we demonstrate that ectopic expression of AtSINA2 in soybean improved the shoot growth, grain yield, drought tolerance and seed oil content in transgenic plants. Compared to wild type, transgenic soybean produced greater shoot biomass and grain yield, and showed improved seed oil and drought tolerance. Physiological analyses exhibited that the increased drought tolerance of transgenic plants was accompanied with a higher chlorophyll content, and a lower malondialdehyde accumulation and water loss during drought stress. Further transcriptomic analyses revealed that the expressions of genes related to plant growth, flowering and stress response were up- or down-regulated in transgenic soybean under both normal and drought stress conditions. Our findings imply that AtSINA2 improved both agricultural production and drought tolerance, and it can be used as a candidate gene for the genetic engineering of new soybean cultivars with improved grain yield and drought resistance.
Collapse
Affiliation(s)
- Jin Yang
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Tingting Mao
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; Zhaoyuan Shenghui Agricultural Technology Development Co, Ltd, North of Beiyuanzhuang Village, Fushan County, Zhaoyuan, Shandong, 265400, China
| | - Zigui Geng
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Wenwen Xue
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Lan Ma
- Shandong Institute of Sericulture, Shandong Academy of Agricultural Sciences, 21 Zhichubei Road, Yantai, 264001, China
| | - Yu Jin
- College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Pan Guo
- College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Zitong Qiu
- College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China
| | - Limin Wang
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; Zhaoyuan Shenghui Agricultural Technology Development Co, Ltd, North of Beiyuanzhuang Village, Fushan County, Zhaoyuan, Shandong, 265400, China
| | - Chunyan Yu
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; Zhaoyuan Shenghui Agricultural Technology Development Co, Ltd, North of Beiyuanzhuang Village, Fushan County, Zhaoyuan, Shandong, 265400, China
| | - Yuting Sheng
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; Zhaoyuan Shenghui Agricultural Technology Development Co, Ltd, North of Beiyuanzhuang Village, Fushan County, Zhaoyuan, Shandong, 265400, China
| | - Juan Zhang
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; College of Agriculture, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China.
| | - Hongxia Zhang
- The Engineering Research Institute of Agriculture and Forestry, Ludong University, 186 Hongqizhong Road, Yantai, 264025, China; Shandong Institute of Sericulture, Shandong Academy of Agricultural Sciences, 21 Zhichubei Road, Yantai, 264001, China; Zhaoyuan Shenghui Agricultural Technology Development Co, Ltd, North of Beiyuanzhuang Village, Fushan County, Zhaoyuan, Shandong, 265400, China.
| |
Collapse
|
6
|
Piedade WP, Famulski JK. E3 ubiquitin ligase-mediated regulation of vertebrate ocular development; new insights into the function of SIAH enzymes. Biochem Soc Trans 2021; 49:327-340. [PMID: 33616626 PMCID: PMC7924998 DOI: 10.1042/bst20200613] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/18/2023]
Abstract
Developmental regulation of the vertebrate visual system has been a focus of investigation for generations as understanding this critical time period has direct implications on our understanding of congenital blinding disease. The majority of studies to date have focused on transcriptional regulation mediated by morphogen gradients and signaling pathways. However, recent studies of post translational regulation during ocular development have shed light on the role of the ubiquitin proteasome system (UPS). This rather ubiquitous yet highly diverse system is well known for regulating protein function and localization as well as stability via targeting for degradation by the 26S proteasome. Work from many model organisms has recently identified UPS activity during various milestones of ocular development including retinal morphogenesis, retinal ganglion cell function as well as photoreceptor homeostasis. In particular work from flies and zebrafish has highlighted the role of the E3 ligase enzyme family, Seven in Absentia Homologue (Siah) during these events. In this review, we summarize the current understanding of UPS activity during Drosophila and vertebrate ocular development, with a major focus on recent findings correlating Siah E3 ligase activity with two major developmental stages of vertebrate ocular development, retinal morphogenesis and photoreceptor specification and survival.
Collapse
|
7
|
Furusawa K, Emoto K. Scrap and Build for Functional Neural Circuits: Spatiotemporal Regulation of Dendrite Degeneration and Regeneration in Neural Development and Disease. Front Cell Neurosci 2021; 14:613320. [PMID: 33505249 PMCID: PMC7829185 DOI: 10.3389/fncel.2020.613320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/04/2020] [Indexed: 01/01/2023] Open
Abstract
Dendrites are cellular structures essential for the integration of neuronal information. These elegant but complex structures are highly patterned across the nervous system but vary tremendously in their size and fine architecture, each designed to best serve specific computations within their networks. Recent in vivo imaging studies reveal that the development of mature dendrite arbors in many cases involves extensive remodeling achieved through a precisely orchestrated interplay of growth, degeneration, and regeneration of dendritic branches. Both degeneration and regeneration of dendritic branches involve precise spatiotemporal regulation for the proper wiring of functional networks. In particular, dendrite degeneration must be targeted in a compartmentalized manner to avoid neuronal death. Dysregulation of these developmental processes, in particular dendrite degeneration, is associated with certain types of pathology, injury, and aging. In this article, we review recent progress in our understanding of dendrite degeneration and regeneration, focusing on molecular and cellular mechanisms underlying spatiotemporal control of dendrite remodeling in neural development. We further discuss how developmental dendrite degeneration and regeneration are molecularly and functionally related to dendrite remodeling in pathology, disease, and aging.
Collapse
Affiliation(s)
- Kotaro Furusawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Kitatani Y, Tezuka A, Hasegawa E, Yanagi S, Togashi K, Tsuji M, Kondo S, Parrish JZ, Emoto K. Drosophila miR-87 promotes dendrite regeneration by targeting the transcriptional repressor Tramtrack69. PLoS Genet 2020; 16:e1008942. [PMID: 32764744 PMCID: PMC7439810 DOI: 10.1371/journal.pgen.1008942] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/19/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
To remodel functional neuronal connectivity, neurons often alter dendrite arbors through elimination and subsequent regeneration of dendritic branches. However, the intrinsic mechanisms underlying this developmentally programmed dendrite regeneration and whether it shares common machinery with injury-induced regeneration remain largely unknown. Drosophila class IV dendrite arborization (C4da) sensory neurons regenerate adult-specific dendrites after eliminating larval dendrites during metamorphosis. Here we show that the microRNA miR-87 is a critical regulator of dendrite regeneration in Drosophila. miR-87 knockout impairs dendrite regeneration after developmentally-programmed pruning, whereas miR-87 overexpression in C4da neurons leads to precocious initiation of dendrite regeneration. Genetic analyses indicate that the transcriptional repressor Tramtrack69 (Ttk69) is a functional target for miR-87-mediated repression as ttk69 expression is increased in miR-87 knockout neurons and reducing ttk69 expression restores dendrite regeneration to mutants lacking miR-87 function. We further show that miR-87 is required for dendrite regeneration after acute injury in the larval stage, providing a mechanistic link between developmentally programmed and injury-induced dendrite regeneration. These findings thus indicate that miR-87 promotes dendrite regrowth during regeneration at least in part through suppressing Ttk69 in Drosophila sensory neurons and suggest that developmental and injury-induced dendrite regeneration share a common intrinsic mechanism to reactivate dendrite growth. Dendrites are the primary sites for synaptic and sensory inputs. To remodel or repair neuronal connectivity, dendrites often exhibit large-scale structural changes that can be triggered by developmental signals, alterations in sensory inputs, or injury. Despite the importance of dendritic remodeling to nervous system function, the molecular basis for this remodeling is largely unknown. Here we used an unbiased genetic screen and in vivo imaging in Drosophila sensory neurons to demonstrate that the microRNA miR-87 is a critical factor required in neurons to reactivate dendritic growth both in developmental remodeling and following injury. Our work supports the model that miR-87 promotes dendrite regeneration by blocking expression of the transcriptional repressor Tramtrack69 in neurons. This study thus establishes a role for miRNAs in temporal control of dendrite regeneration.
Collapse
Affiliation(s)
- Yasuko Kitatani
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Akane Tezuka
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Eri Hasegawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Satoyoshi Yanagi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kazuya Togashi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masato Tsuji
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Yata, Mishima, Shizuoka, Japan
| | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (JZP); (KE)
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- * E-mail: (JZP); (KE)
| |
Collapse
|
9
|
Li HL, Wang X, Ji XL, Qiao ZW, You CX, Hao YJ. Genome-Wide Identification of Apple Ubiquitin SINA E3 Ligase and Functional Characterization of MdSINA2. FRONTIERS IN PLANT SCIENCE 2020; 11:1109. [PMID: 32793265 PMCID: PMC7393226 DOI: 10.3389/fpls.2020.01109] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/06/2020] [Indexed: 05/22/2023]
Abstract
SINA (Seven in absentia) proteins are a small family of ubiquitin ligases that play important roles in regulating plant growth and developmental processes as well as in responses to diverse types of biotic and abiotic stress. However, the characteristics of the apple SINA family have not been previously studied. Here, we identified 11 MdSINAs members in the apple genome based on their conserved, N-terminal RING and C-terminal SINA domains. We also reconstructed a phylogeny of these genes; characterized their chromosomal location, structure, and motifs; and identified two major groups of MdSINA genes. Subsequent qRT-PCR analyses were used to characterize the expression of MdSINA genes in various tissues and organs, and levels of expression were highest in leaves. MdSINAs were significantly induced under ABA and carbon- and nitrate-starvation treatment. Except for MdSINA1 and MdSINA7, the other MdSINA proteins could interact with each other. Moreover, MdSINA2 was found to be localized in the nucleus using Agrobacterium-mediated transient expression. Western-blot analysis showed that MdSINA2 accumulated extensively under light, decreased under darkness, and became insensitive to light when the RING domain was disrupted. Finally, ABA-hypersensitive phenotypes were confirmed by transgenic calli and the ectopic expression of MdSINA2 in Arabidopsis. In conclusion, our results suggest that MdSINA genes participate in the responses to different types of stress, and that MdSINA2 might act as a negative regulator in the ABA stress response.
Collapse
|
10
|
Hughes SE, Hemenway E, Guo F, Yi K, Yu Z, Hawley RS. The E3 ubiquitin ligase Sina regulates the assembly and disassembly of the synaptonemal complex in Drosophila females. PLoS Genet 2019; 15:e1008161. [PMID: 31107865 PMCID: PMC6544331 DOI: 10.1371/journal.pgen.1008161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/31/2019] [Accepted: 04/26/2019] [Indexed: 01/04/2023] Open
Abstract
During early meiotic prophase, homologous chromosomes are connected along their entire lengths by a proteinaceous tripartite structure known as the synaptonemal complex (SC). Although the components that comprise the SC are predominantly studied in this canonical ribbon-like structure, they can also polymerize into repeated structures known as polycomplexes. We find that in Drosophila oocytes, the ability of SC components to assemble into canonical tripartite SC requires the E3 ubiquitin ligase Seven in absentia (Sina). In sina mutant oocytes, SC components assemble into large rod-like polycomplexes instead of proper SC. Thus, the wild-type Sina protein inhibits the polymerization of SC components, including those of the lateral element, into polycomplexes. These polycomplexes persist into meiotic stages when canonical SC has been disassembled, indicating that Sina also plays a role in controlling SC disassembly. Polycomplexes induced by loss-of-function sina mutations associate with centromeres, sites of double-strand breaks, and cohesins. Perhaps as a consequence of these associations, centromere clustering is defective and crossing over is reduced. These results suggest that while features of the polycomplexes can be recognized as SC by other components of the meiotic nucleus, polycomplexes nonetheless fail to execute core functions of canonical SC.
Collapse
Affiliation(s)
- Stacie E. Hughes
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Elizabeth Hemenway
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Fengli Guo
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - R. Scott Hawley
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
11
|
Sirover MA. Pleiotropic effects of moonlighting glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in cancer progression, invasiveness, and metastases. Cancer Metastasis Rev 2019; 37:665-676. [PMID: 30209795 DOI: 10.1007/s10555-018-9764-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) may represent the quintessential example of a moonlighting protein. The latter are a new, intriguing class of cell proteins which exhibit multiple activities in different subcellular locales apart from their initially, well-characterized function. As such, apart from its classical role in energy production, membrane-bound GAPDH is required for membrane fusion, endocytosis and, intriguingly, for iron transport. Cytoplasmic GAPDH regulates mRNA stability and is required for ER to Golgi trafficking. Nuclear GAPDH is involved in apoptosis, transcriptional gene regulation, the maintenance of DNA integrity, as well as nuclear tRNA export. Paradoxically, the etiology of a number of human pathologies is dependent upon GAPDH structure and function. In particular, recent evidence indicates a significant role for moonlighting GAPDH in tumorigenesis. Specifically, these include its role in the survival of tumor cells, in tumor angiogenesis, as well as its control of tumor cell gene expression and posttranscriptional regulation of tumor cell mRNA. Each of these activities correlates with increased tumor progression and, unfortunately, a poor prognosis for the afflicted individual.
Collapse
Affiliation(s)
- Michael A Sirover
- Department of Pharmacology, The Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19047, USA.
| |
Collapse
|
12
|
Zhou Y, Popadowski SE, Deutschman E, Halfon MS. Distinct roles and requirements for Ras pathway signaling in visceral versus somatic muscle founder specification. Development 2019; 146:dev.169003. [PMID: 30630823 DOI: 10.1242/dev.169003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Pleiotropic signaling pathways must somehow engender specific cellular responses. In the Drosophila mesoderm, Ras pathway signaling specifies muscle founder cells from among the broader population of myoblasts. For somatic muscles, this is an inductive process mediated by the ETS-domain downstream Ras effectors Pointed and Aop (Yan). We demonstrate here that for the circular visceral muscles, despite superficial similarities, a significantly different specification mechanism is at work. Not only is visceral founder cell specification not dependent on Pointed or Aop, but Ras pathway signaling in its entirety can be bypassed. Our results show that de-repression, not activation, is the predominant role of Ras signaling in the visceral mesoderm and that, accordingly, Ras signaling is not required in the absence of repression. The key repressor acts downstream of the transcription factor Lame duck and is likely a member of the ETS transcription factor family. Our findings fit with a growing body of data that point to a complex interplay between the Ras pathway, ETS transcription factors, and enhancer binding as a crucial mechanism for determining unique responses to Ras signaling.
Collapse
Affiliation(s)
- Yiyun Zhou
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
| | - Sarah E Popadowski
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
| | - Emily Deutschman
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA
| | - Marc S Halfon
- Department of Biochemistry, University at Buffalo-State University of New York, Buffalo, NY 14203, USA .,Department of Biological Sciences, University at Buffalo-State University of New York, Buffalo, NY 14203, USA.,Department of Biomedical Informatics, University at Buffalo-State University of New York, Buffalo, NY 14203, USA.,NY State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.,Molecular and Cellular Biology Department and Program in Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
13
|
Yin C, Xi R. A Phyllopod-Mediated Feedback Loop Promotes Intestinal Stem Cell Enteroendocrine Commitment in Drosophila. Stem Cell Reports 2017; 10:43-57. [PMID: 29276156 PMCID: PMC5768918 DOI: 10.1016/j.stemcr.2017.11.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 01/17/2023] Open
Abstract
The intestinal epithelium in the Drosophila midgut is maintained by intestinal stem cells (ISCs), which are capable of generating both enterocytes and enteroendocrine cells (EEs) via alternative cell fate specification. Activation of Delta-Notch signaling directs ISCs for enterocyte generation, but how EEs are generated from ISCs remains poorly understood. Here, we identified Phyllopod (Phyl) as a key regulator that drives EE generation from ISCs. Phyl, which is normally suppressed by Notch, functions as an adaptor protein that bridges Tramtrack 69 (Ttk69) and E3 ubiquitin ligase Sina for degradation. Degradation of Ttk69 allows the activation of the Achaete-Scute Complex (AS-C)-Pros regulatory axis, which promotes EE specification. Interestingly, expression of AS-C genes in turn further induces Phyl expression, thereby establishing a positive feedback loop for continuous EE fate specification and commitment. This positive feedback circuit-driven regulatory mechanism could represent a common strategy for reliable and irreversible cell fate determination from progenitor cells.
Collapse
Affiliation(s)
- Chang Yin
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Rongwen Xi
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China; Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| |
Collapse
|
14
|
Ong T, Solecki DJ. Seven in Absentia E3 Ubiquitin Ligases: Central Regulators of Neural Cell Fate and Neuronal Polarity. Front Cell Neurosci 2017; 11:322. [PMID: 29081737 PMCID: PMC5646344 DOI: 10.3389/fncel.2017.00322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/31/2022] Open
Abstract
During neural development, neural precursors transition from a proliferative state within their germinal niches to a migratory state as they relocate to their final laminar positions. Transitions across these states are coupled with dynamic alterations in cellular polarity. This key feature can be seen throughout the developing vertebrate brain, in which neural stem cells give rise to multipolar or unpolarized transit-amplifying progenitors. These transit-amplifying progenitors then expand to give rise to mature neuronal lineages that become polarized as they initiate radial migration to their final laminar positions. The conventional understanding of the cellular polarity regulatory program has revolved around signaling cascades and transcriptional networks. In this review, we discuss recent discoveries concerning the role of the Siah2 ubiquitin ligase in initiating neuronal polarity during cerebellar development. Given the unique features of Siah ubiquitin ligases, we highlight some of the key substrates that play important roles in cellular polarity and propose a function for the Siah ubiquitin proteasome pathway in mediating a post-translational regulatory network to control the onset of polarization.
Collapse
Affiliation(s)
- Taren Ong
- Cancer and Developmental Biology Track, Integrated Biomedical Sciences Graduate Program, University of Tennessee Health Science Center, Memphis, TN, United States.,Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
15
|
Mavromatakis YE, Tomlinson A. R7 Photoreceptor Specification in the Developing Drosophila Eye: The Role of the Transcription Factor Deadpan. PLoS Genet 2016; 12:e1006159. [PMID: 27427987 PMCID: PMC4948816 DOI: 10.1371/journal.pgen.1006159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/10/2016] [Indexed: 02/02/2023] Open
Abstract
As cells proceed along their developmental pathways they make a series of sequential cell fate decisions. Each of those decisions needs to be made in a robust manner so there is no ambiguity in the state of the cell as it proceeds to the next stage. Here we examine the decision made by the Drosophila R7 precursor cell to become a photoreceptor and ask how the robustness of that decision is achieved. The transcription factor Tramtrack (Ttk) inhibits photoreceptor assignment, and previous studies found that the RTK-induced degradation of Ttk was critically required for R7 specification. Here we find that the transcription factor Deadpan (Dpn) is also required; it is needed to silence ttk transcription, and only when Ttk protein degradation and transcriptional silencing occur together is the photoreceptor fate robustly achieved. Dpn expression needs to be tightly restricted to R7 precursors, and we describe the role played by Ttk in repressing dpn transcription. Thus, Dpn and Ttk act as mutually repressive transcription factors, with Dpn acting to ensure that Ttk is effectively removed from R7, and Ttk acting to prevent Dpn expression in other cells. Furthermore, we find that N activity is required to promote dpn transcription, and only in R7 precursors does the removal of Ttk coincide with high N activity, and only in this cell does Dpn expression result. Animals are made from a vast diversity of different cell types, and understanding how they are specified is a major goal of developmental biology. In this study we use the Drosophila R7 photoreceptor as a model system for understanding how cell fate specification occurs. We examine the step when the R7 precursor cell adopts the photoreceptor fate, and ask how the signaling pathways active in the cell are integrated to provide an unambiguous directive to become a photoreceptor. The transcription factor Tramtrack (Ttk) represses the ability of the cell to become a photoreceptor, and how it is removed is the focus of this study. Previous work identified a protein degradation mechanism, and here we describe the role of the transcription factor Deadpan (Dpn) in repressing ttk transcription. We find that both the protein degradation mechanism and transcriptional silencing are required for efficient Ttk removal. Dpn expression needs to be restricted to the R7 precursor and we describe how the mutual antagonism between Ttk and Dpn and the action of the Notch signaling pathway are integrated to ensure that Dpn is selectively expressed in the cell.
Collapse
Affiliation(s)
- Yannis Emmanuel Mavromatakis
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| | - Andrew Tomlinson
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
Peralta DA, Araya A, Busi MV, Gomez-Casati DF. The E3 ubiquitin-ligase SEVEN IN ABSENTIA like 7 mono-ubiquitinates glyceraldehyde-3-phosphate dehydrogenase 1 isoform in vitro and is required for its nuclear localization in Arabidopsis thaliana. Int J Biochem Cell Biol 2015; 70:48-56. [PMID: 26582368 DOI: 10.1016/j.biocel.2015.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/28/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
The E3 ubiquitin-protein ligases are associated to various processes such as cell cycle control and diverse developmental pathways. Arabidopsis thaliana SEVEN IN ABSENTIA like 7, which has ubiquitin ligase activity, is located in the nucleus and cytosol and is expressed at several stages in almost all plant tissues suggesting an important role in plant functions. However, the mechanism underlying the regulation of this protein is unknown. Since we found that the SEVEN IN ABSENTIA like 7 gene expression is altered in plants with impaired mitochondria, and in plants deficient in the glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase 1, we decided to study the possible interactions between both proteins as potential partners in plant signaling functions. We found that SEVEN IN ABSENTIA like 7 is able to interact in vitro with glyceraldehyde-3-phosphate dehydrogenase and that the Lys231 residue of the last is essential for this function. Following the interaction, a concomitant increase in the glyceraldehyde-3-phosphate dehydrogenase catalytic activity was observed. However, when SEVEN IN ABSENTIA like 7 was supplemented with E1 and E2 proteins to form a complete E1-E2-E3 modifier complex, we observed the mono-ubiquitination of glyceraldehyde-3-phosphate dehydrogenase 1 at the Lys76 residue and a dramatic decrease of its catalytic activity. Moreover, we found that localization of glyceraldehyde-3-phosphate dehydrogenase 1 in the nucleus is dependent on the expression SEVEN IN ABSENTIA like 7. These observations suggest that the association of both proteins might result in different biological consequences in plants either through affecting the glycolytic flux or via cytoplasm-nucleus relocation.
Collapse
Affiliation(s)
- Diego A Peralta
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina.
| | - Alejandro Araya
- Centre National de la Recherche Scientifique & UMR 1332 - Biologie du Fruit et Pathologie, Institute National de la Recherche Agronomique (INRA) Bordeaux Aquitaine, 71 avenue Edouard Bourlaux, Villenave D'Ornon 33882, France.
| | - Maria V Busi
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina.
| | - Diego F Gomez-Casati
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Suipacha 531, Rosario 2000, Argentina.
| |
Collapse
|
17
|
Wang C, Guo X, Dou K, Chen H, Xi R. Ttk69 acts as a master repressor of enteroendocrine cell specification in Drosophila intestinal stem cell lineages. Development 2015; 142:3321-31. [PMID: 26293304 DOI: 10.1242/dev.123208] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/10/2015] [Indexed: 01/15/2023]
Abstract
In adult Drosophila midgut, intestinal stem cells (ISCs) periodically produce progenitor cells that undergo a binary fate choice determined primarily by the levels of Notch activity that they receive, before terminally differentiating into enterocytes (ECs) or enteroendocrine (EE) cells. Here we identified Ttk69, a BTB domain-containing transcriptional repressor, as a master repressor of EE cell specification in the ISC lineages. Depletion of ttk69 in progenitor cells induced ISC proliferation and caused all committed progenitor cells to adopt EE fate, leading to the production of supernumerary EE cells in the intestinal epithelium. Conversely, forced expression of Ttk69 in progenitor cells was sufficient to prevent EE cell specification. The expression of Ttk69 was not regulated by Notch signaling, and forced activation of Notch, which is sufficient to induce EC specification of normal progenitor cells, failed to prevent EE cell specification of Ttk69-depleted progenitors. Loss of Ttk69 led to derepression of the acheate-scute complex (AS-C) genes scute and asense, which then induced prospero expression to promote EE cell specification. These studies suggest that Ttk69 functions in parallel with Notch signaling and acts as a master repressor of EE cell specification in Drosophila ISC lineages primarily by suppressing AS-C genes.
Collapse
Affiliation(s)
- Chenhui Wang
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Xingting Guo
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Kun Dou
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Hongyan Chen
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| | - Rongwen Xi
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, China
| |
Collapse
|
18
|
Ciglar L, Girardot C, Wilczyński B, Braun M, Furlong EEM. Coordinated repression and activation of two transcriptional programs stabilizes cell fate during myogenesis. Development 2014; 141:2633-43. [PMID: 24961800 PMCID: PMC4146391 DOI: 10.1242/dev.101956] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Molecular models of cell fate specification typically focus on the activation of specific lineage programs. However, the concurrent repression of unwanted transcriptional networks is also essential to stabilize certain cellular identities, as shown in a number of diverse systems and phyla. Here, we demonstrate that this dual requirement also holds true in the context of Drosophila myogenesis. By integrating genetics and genomics, we identified a new role for the pleiotropic transcriptional repressor Tramtrack69 in myoblast specification. Drosophila muscles are formed through the fusion of two discrete cell types: founder cells (FCs) and fusion-competent myoblasts (FCMs). When tramtrack69 is removed, FCMs appear to adopt an alternative muscle FC-like fate. Conversely, ectopic expression of this repressor phenocopies muscle defects seen in loss-of-function lame duck mutants, a transcription factor specific to FCMs. This occurs through Tramtrack69-mediated repression in FCMs, whereas Lame duck activates a largely distinct transcriptional program in the same cells. Lineage-specific factors are therefore not sufficient to maintain FCM identity. Instead, their identity appears more plastic, requiring the combination of instructive repressive and activating programs to stabilize cell fate.
Collapse
Affiliation(s)
- Lucia Ciglar
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Charles Girardot
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Bartek Wilczyński
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Martina Braun
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Eileen E M Furlong
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| |
Collapse
|
19
|
Elgart M, Snir O, Soen Y. Stress-mediated tuning of developmental robustness and plasticity in flies. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:462-6. [PMID: 25134463 DOI: 10.1016/j.bbagrm.2014.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 11/30/2022]
Abstract
Organisms have to be sufficiently robust to environmental and genetic perturbations, yet plastic enough to cope with stressful scenarios to which they are not fully adapted. How this apparent conflict between robustness and plasticity is resolved at the cellular and whole organism levels is not clear. Here we review and discuss evidence in flies suggesting that the environment can modulate the balance between robustness and plasticity. The outcomes of this modulation can vary from mild sensitizations that are hardly noticeable, to overt qualitative changes in phenotype. The effects could be at both the cellular and whole organism levels and can include cellular de-/trans-differentiation ('Cellular reprogramming') and gross disfigurements such as homeotic transformations ('Tissue/whole organism reprogramming'). When the stress is mild enough, plastic changes in some processes may prevent drastic changes in more robust traits such as cell identity and tissue integrity. However, when the stress is sufficiently severe, this buffering may no longer be able to prevent such overt changes, and the resulting phenotypic variability could be subjected to selection and might assist survival at the population level. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.
Collapse
Affiliation(s)
- M Elgart
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - O Snir
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Y Soen
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
20
|
Kang N, Won M, Rhee M, Ro H. Siah ubiquitin ligases modulate nodal signaling during zebrafish embryonic development. Mol Cells 2014; 37:389-98. [PMID: 24823357 PMCID: PMC4044310 DOI: 10.14348/molcells.2014.0032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/27/2014] [Accepted: 03/31/2014] [Indexed: 01/15/2023] Open
Abstract
Siah2 is a zebrafish homologue of mammalian Siah family. Siah acts as an E3 ubiquitin ligase that binds proteins destined for degradation. Extensive homology between siah and Drosophila Siah homologue (sina) suggests their important physiological roles during embryonic development. However, detailed functional studies of Siah in vertebrate development have not been carried out. Here we report that Siah2 specifically augments nodal related gene expression in marginal blastomeres at late blastula through early gastrula stages of zebrafish embryos. Siah2 dependent Nodal signaling augmentation is confirmed by cell-based reporter gene assays using 293T cells and 3TPluciferase reporter plasmid. We also established a molecular hierarchy of Siah as a upstream regulator of FoxH1/Fast1 transcriptional factor in Nodal signaling. Elevated expression of nodal related genes by overexpression of Siah2 was enough to override the inhibitory effects of atv and lft2 on the Nodal signaling. In particular, E3 ubiquitin ligase activity of Siah2 is critical to limit the duration and/or magnitude of Nodal signaling. Additionally, since the embryos injected with Siah morpholinos mimicked the atv overexpression phenotype at least in part, our data support a model in which Siah is involved in mesendoderm patterning via modulating Nodal signaling.
Collapse
Affiliation(s)
- Nami Kang
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764,
Korea
| | - Minho Won
- Program in Genomics of Differentiation, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland,
USA
| | - Myungchull Rhee
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764,
Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764,
Korea
| |
Collapse
|
21
|
Abstract
The developing Drosophila ommatidium is characterized by two distinct waves of pattern formation. In the first wave, a precluster of five cells is formed by a complex cellular interaction mechanism. In the second wave, cells are systematically recruited to the cluster and directed to their fates by developmental cues presented by differentiating precluster cells. These developmental cues are mediated through the receptor tyrosine kinase (RTK) and Notch (N) signaling pathways and their combined activities are crucial in specifying cell type. The transcription factor Lozenge (Lz) is expressed exclusively in second wave cells. Here, we ectopically supply Lz to precluster cells and concomitantly supply the various RTK/N codes that specify each of three second wave cell fates. We thereby reproduce molecular markers of each of the second wave cell types in precluster cells and draw three inferences. First, we confirm that Lz provides key intrinsic information to second wave cells. We can now combine this with the RTK/N signaling to provide a cell fate specification code that entails both extrinsic and intrinsic information. Second, the reproduction of each second wave cell type in the precluster confirms the accuracy of the RTK/N signaling code. Third, RTK/N signaling and Lz need only be presented to the cells for a short period of time in order to specify their fate.
Collapse
Affiliation(s)
- Yannis Emmanuel Mavromatakis
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
22
|
Peralta DA, Araya A, Nardi CF, Busi MV, Gomez-Casati DF. Characterization of the Arabidopsis thaliana E3 ubiquitin-ligase AtSINAL7 and identification of the ubiquitination sites. PLoS One 2013; 8:e73104. [PMID: 24015288 PMCID: PMC3756039 DOI: 10.1371/journal.pone.0073104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 07/25/2013] [Indexed: 12/05/2022] Open
Abstract
Protein ubiquitination leading to degradation by the proteasome is an important mechanism in regulating key cellular functions. Protein ubiquitination is carried out by a three step process involving ubiquitin (Ub) activation by a E1 enzyme, the transfer of Ub to a protein E2, finally an ubiquitin ligase E3 catalyzes the transfer of the Ub peptide to an acceptor protein. The E3 component is responsible for the specific recognition of the target, making the unveiling of E3 components essential to understand the mechanisms regulating fundamental cell processes through the protein degradation pathways. The Arabidopsis thaliana seven in absentia-like 7 (AtSINAL7) gene encodes for a protein with characteristics from a C3HC4-type E3 ubiquitin ligase. We demonstrate here that AtSINAL7 protein is indeed an E3 protein ligase based on the self-ubiquitination in vitro assay. This activity is dependent of the presence of a Lys residue in position 124. We also found that higher AtSINAL7 transcript levels are present in tissues undergoing active cell division during floral development. An interesting observation is the circadian expression pattern of AtSINAL7 mRNA in floral buds. Furthermore, UV–B irradiation induces the expression of this transcript indicating that AtSINAL7 may be involved in a wide range of different cell processes.
Collapse
Affiliation(s)
- Diego A. Peralta
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
| | - Alejandro Araya
- Centre National de la Recherche Scientifique and UMR 1332 – Biologie du Fruit et Pathologie, Institute National de la Recherche Agronomique (INRA) Bordeaux Aquitaine, Villenave D’Ornon, France
| | - Cristina F. Nardi
- Instituto de Investigaciones Biotecnológicas, Instituto Tecnológico de Chascomús (IIB-INTECH-CONICET), Universidad Nacional de San Martin, Chascomús, Argentina
| | - Maria V. Busi
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- * E-mail: (MVB); (DFG-C)
| | - Diego F. Gomez-Casati
- Centro de Estudios Fotosintéticos y Bioquímicos (CEFOBI-CONICET), Universidad Nacional de Rosario, Rosario, Argentina
- * E-mail: (MVB); (DFG-C)
| |
Collapse
|
23
|
Stebbins JL, Santelli E, Feng Y, De SK, Purves A, Motamedchaboki K, Wu B, Ronai ZA, Liddington RC, Pellecchia M. Structure-based design of covalent Siah inhibitors. ACTA ACUST UNITED AC 2013; 20:973-82. [PMID: 23891150 DOI: 10.1016/j.chembiol.2013.06.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/13/2013] [Accepted: 06/17/2013] [Indexed: 11/30/2022]
Abstract
The E3 ubiquitin ligase Siah regulates key cellular events that are central to cancer development and progression. A promising route to Siah inhibition is disrupting its interactions with adaptor proteins. However, typical of protein-protein interactions, traditional unbiased approaches to ligand discovery did not produce viable hits against this target, despite considerable effort and a multitude of approaches. Ultimately, a rational structure-based design strategy was successful for the identification of Siah inhibitors in which peptide binding drives specific covalent bond formation with the target. X-ray crystallography, mass spectrometry, and functional data demonstrate that these peptide mimetics are efficient covalent inhibitors of Siah and antagonize Siah-dependent regulation of Erk and Hif signaling in the cell. The proposed strategy may result useful as a general approach to the design of peptide-based inhibitors of other protein-protein interactions.
Collapse
Affiliation(s)
- John L Stebbins
- Signal Transduction Program and Cell Death Program, Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Siah2 regulates tight junction integrity and cell polarity through control of ASPP2 stability. Oncogene 2013; 33:2004-10. [PMID: 23644657 PMCID: PMC3917971 DOI: 10.1038/onc.2013.149] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 01/21/2013] [Accepted: 03/12/2013] [Indexed: 12/24/2022]
Abstract
Changes in cell adhesion and polarity are closely associated with epithelial cell transformation and metastatic capacity. The tumor suppressor protein ASPP2 has been implicated in control of cell adhesion and polarity, through its effect on the PAR complex. Here we demonstrate that under hypoxic conditions the ubiquitin ligase Siah2 controls ASPP2 availability, with concomitant effect on epithelial cell polarity. LC-MS/MS analysis identified ASPP2 and ASPP1 as Siah2 interacting proteins. Biochemical analysis confirmed this interaction and mapped degron motifs within ASPP2, which are required for Siah2-mediated ubiquitination and proteasomal-dependent degradation. Inhibition of Siah2 expression increases ASPP2 levels and enhances ASPP2-dependent maintenance of TJ integrity and polarized architecture in 3D organotypic culture. Conversely, increase of Siah2 expression under hypoxia decreases ASPP2 levels and the formation of apical polarity in 3D culture. In all, our studies demonstrate the role of Siah2 in regulation of TJ integrity and cell polarity under hypoxia, through its regulation of ASPP2 stability.
Collapse
|
25
|
R7 photoreceptor axon growth is temporally controlled by the transcription factor Ttk69, which inhibits growth in part by promoting transforming growth factor-β/activin signaling. J Neurosci 2013; 33:1509-20. [PMID: 23345225 DOI: 10.1523/jneurosci.2023-12.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Work on axon growth has classically focused on understanding how extrinsic cues control growth cone dynamics independent of the cell body. However, more recently, neuron-intrinsic transcription factors have been shown to influence both normal and regenerative axon growth, suggesting that understanding their mechanism of action is of clinical importance. We are studying axon targeting in the Drosophila visual system and here show that the BTB/POZ zinc-finger transcription factor Tramtrack69 (Ttk69) plays an instructive role in inhibiting the growth of R7 photoreceptor axon terminals. Although ttk69 mutant R7 axons project to the correct medullar target layer, M6, their terminals fail to remain retinotopically restricted and instead grow laterally within M6. This overgrowth is not caused by an inability to be repelled by neighboring R7 axons or by an inability to recognize and initiate synapse formation with postsynaptic targets. The overgrowth is progressive and occurs even if contact between ttk69 mutant R7 axons and their normal target layer is disrupted. Ttk69 is first expressed in wild-type R7s after their axons have reached the medulla; ttk69 mutant R7 axon terminal overgrowth begins shortly after this time point. We find that expressing Ttk69 prematurely in R7s collapses their growth cones and disrupts axon extension, indicating that Ttk69 plays an instructive role in this process. A TGF-β/Activin pathway was shown previously to inhibit R7 axon terminal growth. We find that Ttk69 is required for normal activation of this pathway but that Ttk69 likely also inhibits R7 axon growth by a TGF-β/Activin-independent mechanism.
Collapse
|
26
|
Treisman JE. Retinal differentiation in Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:545-57. [PMID: 24014422 DOI: 10.1002/wdev.100] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drosophila eye development has been extensively studied, due to the ease of genetic screens for mutations disrupting this process. The eye imaginal disc is specified during embryonic and larval development by the Pax6 homolog Eyeless and a network of downstream transcription factors. Expression of these factors is regulated by signaling molecules and also indirectly by growth of the eye disc. Differentiation of photoreceptor clusters initiates in the third larval instar at the posterior of the eye disc and progresses anteriorly, driven by the secreted protein Hedgehog. Within each cluster, the combined activities of Hedgehog signaling and Notch-mediated lateral inhibition induce and refine the expression of the transcription factor Atonal, which specifies the founding R8 photoreceptor of each ommatidium. Seven additional photoreceptors, followed by cone and pigment cells, are successively recruited by the signaling molecules Spitz, Delta, and Bride of sevenless. Combinations of these signals and of intrinsic transcription factors give each ommatidial cell its specific identity. During the pupal stages, rhodopsins are expressed, and the photoreceptors and accessory cells take on their final positions and morphologies to form the adult retina. Over the past few decades, the genetic analysis of this small number of cell types arranged in a repetitive structure has allowed a remarkably detailed understanding of the basic mechanisms controlling cell differentiation and morphological rearrangement.
Collapse
Affiliation(s)
- Jessica E Treisman
- Department of Cell Biology and Kimmel Center for Biology and Medicine of the Skirball Institute, NYU School of Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Pérez M, García-Limones C, Zapico I, Marina A, Schmitz ML, Muñoz E, Calzado MA. Mutual regulation between SIAH2 and DYRK2 controls hypoxic and genotoxic signaling pathways. J Mol Cell Biol 2012; 4:316-30. [PMID: 22878263 DOI: 10.1093/jmcb/mjs047] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The ubiquitin E3 ligase SIAH2 is an important regulator of the hypoxic response as it leads to the ubiquitin/proteasomal degradation of prolyl hydroxylases such as PHD3, which in turn increases the stability of hypoxia-inducible factor (HIF)-1α. In the present study, we identify the serine/threonine kinase DYRK2 as SIAH2 interaction partner that phosphorylates SIAH2 at five residues (Ser16, Thr26, Ser28, Ser68, and Thr119). Phosphomimetic and phospho-mutant forms of SIAH2 exhibit different subcellular localizations and consequently change in PHD3 degrading activity. Accordingly, phosphorylated SIAH2 is more active than the wild-type E3 ligase and shows an increased ability to trigger the HIF-1α-mediated transcriptional response and angiogenesis. We also found that SIAH2 knockdown increases DYRK2 stability, whereas SIAH2 expression facilitates DYRK2 polyubiquitination and degradation. Hypoxic conditions cause a SIAH2-dependent DYRK2 polyubiquitination and degradation which ultimately also results in an impaired SIAH2 phosphorylation. Similarly, DYRK2-mediated phosphorylation of p53 at Ser46 is impaired under hypoxic conditions, suggesting a molecular mechanism underlying chemotherapy resistance in solid tumors.
Collapse
Affiliation(s)
- Moisés Pérez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Universidad de Córdoba, 14004 Córdoba, Spain
| | | | | | | | | | | | | |
Collapse
|
28
|
Mavromatakis YE, Tomlinson A. Stop and go: antagonistic signals in the specification of the Drosophila R7 photoreceptor viewed from an evolutionary perspective. Fly (Austin) 2012; 6:228-33. [PMID: 22878552 DOI: 10.4161/fly.21102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The Drosophila R7 photoreceptor precursor is directed to its fate by signals from adjacent cells that activate its Receptor Tyrosine Kinase (RTK) and Notch (N) signaling pathways. Counter-intuitively, the N activity both promotes and inhibits the photoreceptor fate in the R7 precursor. We offer an evolutionary perspective for this in which earlier ommatidia had fewer photoreceptors and used N to inhibit the addition of any more. When additional photoreceptors were added by evolution, an RTK signal was used to overcome the N inhibition in these cells, and these new additions potently activated N in their neighboring cells, preventing them from also responding to the RTK signal. The R7 precursor also receives this block, and requires robust RTK activation for it to become a photoreceptor. This is achieved by N transcriptionally activating a new RTK, one that is potently activated in the R7 precursor and sufficing to overcome the N inhibition. The unusually high RTK signal in R7 requires additional transduction components not needed when the signal is mild; in R7 the small GTPases Ras and Rap are both required to transduce the signal, but in other photoreceptors Ras alone suffices.
Collapse
|
29
|
Abstract
Since the discovery of a single white-eyed male in a population of red eyed flies over 100 years ago (Morgan, 1910), the compound eye of the fruit fly, Drosophila melanogaster, has been a favorite experimental system for identifying genes that regulate various aspects of development. For example, a fair amount of what we know today about enzymatic pathways and vesicular transport is due to the discovery and subsequent characterization of eye color mutants such as white. Likewise, our present day understanding of organogenesis has been aided considerably by studies of mutations, such as eyeless, that either reduce or eliminate the compound eyes. But by far the phenotype that has provided levers into the greatest number of experimental fields has been the humble "rough" eye. The fly eye is composed of several hundred unit-eyes that are also called ommatidia. These unit eyes are packed into a hexagonal array of remarkable precision. The structure of the eye is so precise that it has been compared with that of a crystal (Ready et al., 1976). Even the slightest perturbations to the structure of the ommatidium can be visually detected by light or electron microscopy. The cause for this is two-fold: (1) any defect that affects the hexagonal geometry of a single ommatidium can and will disrupt the positioning of surrounding unit eyes thereby propagating structural flaws and (2) disruptions in genes that govern the development of even a single cell within an ommatidium will affect all unit eyes. In both cases, the effect is the visual magnification of even the smallest imperfection. Studies of rough eye mutants have provided key insights into the areas of cell fate specification, lateral inhibition, signal transduction, transcription factor networks, planar cell polarity, cell proliferation, and programmed cell death just to name a few. This review will attempt to summarize the key steps that are required to assemble each ommatidium.
Collapse
Affiliation(s)
- Justin P Kumar
- Department of Biology, Indiana University, Bloomington, Indiana 47405, USA.
| |
Collapse
|
30
|
Casad ME, Yu L, Daniels JP, Wolf MJ, Rockman HA. Deletion of Siah-interacting protein gene in Drosophila causes cardiomyopathy. Mol Genet Genomics 2012; 287:351-60. [PMID: 22398840 DOI: 10.1007/s00438-012-0684-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/22/2012] [Indexed: 01/18/2023]
Abstract
Drosophila is a useful model organism in which the genetics of human diseases, including recent advances in identification of the genetics of heart development and disease in the fly, can be studied. To identify novel genes that cause cardiomyopathy, we performed a deficiency screen in adult Drosophila. Using optical coherence tomography to phenotype cardiac function in awake adult Drosophila, we identified Df(1)Exel6240 as having cardiomyopathy. Using a number of strategies including customized smaller deletions, screening of mutant alleles, and transgenic rescue, we identified CG3226 as the causative gene for this deficiency. CG3226 is an uncharacterized gene in Drosophila possessing homology to the mammalian Siah-interacting protein (SIP) gene. Mammalian SIP functions as an adaptor protein involved in one of the β-catenin degradation complexes. To investigate the effects of altering β-catenin/Armadillo signaling in the adult fly, we measured heart function in flies expressing either constitutively active Armadillo or transgenic constructs that block Armadillo signaling, specifically in the heart. While, increasing Armadillo signaling in the heart did not have an effect on adult heart function, decreasing Armadillo signaling in the fly heart caused the significant reduction in heart chamber size. In summary, we show that deletion of CG3226, which has homology to mammalian SIP, causes cardiomyopathy in adult Drosophila. Alterations in Armadillo signaling during development lead to important changes in the size and function of the adult heart.
Collapse
Affiliation(s)
- Michelle E Casad
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
31
|
The role of the small GTPase Rap in Drosophila R7 photoreceptor specification. Proc Natl Acad Sci U S A 2012; 109:3844-9. [PMID: 22355117 DOI: 10.1073/pnas.1115108109] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Drosophila R7 photoreceptor provides an excellent model system with which to study how cells receive and "decode" signals that specify cell fate. R7 is specified by the combined actions of the receptor tyrosine kinase (RTK) and Notch (N) signaling pathways. These pathways interact in a complex manner that includes antagonistic effects on photoreceptor specification: RTK promotes the photoreceptor fate, whereas N inhibits. Although other photoreceptors are subject to only mild N activation, R7 experiences a high-level N signal. To counter this effect and to ensure that the cell is specified as a photoreceptor, a high RTK signal is transduced in the cell. Thus, there are two levels of RTK transduction in the photoreceptors: in R7 it is high, whereas in others it is low. Here, we address how this high-level RTK signal is transduced in R7 and find that, in addition to Ras, another small GTPase, Rap, is also engaged. Thus, when N activity is high, a robust RTK signal operates that uses both Ras and Rap, but when N activity is low, only a mild RTK signal is transduced and Ras alone suffices for the purpose.
Collapse
|
32
|
Wu X, Oh MH, Kim HS, Schwartz D, Imai BS, Yau PM, Clouse SD, Huber SC. Transphosphorylation of E. coli Proteins during Production of Recombinant Protein Kinases Provides a Robust System to Characterize Kinase Specificity. FRONTIERS IN PLANT SCIENCE 2012; 3:262. [PMID: 23226150 PMCID: PMC3510383 DOI: 10.3389/fpls.2012.00262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 11/10/2012] [Indexed: 05/09/2023]
Abstract
Protein kinase specificity is of fundamental importance to pathway regulation and signal transduction. Here, we report a convenient system to monitor the activity and specificity of recombinant protein kinases expressed in E. coli. We apply this to the study of the cytoplasmic domain of the plant receptor kinase BRASSINOSTEROID-INSENSITIVE 1 (BRI1), which functions in brassinosteroid (BR) signaling. Recombinant BRI1 is catalytically active and both autophosphorylates and transphosphorylates E. coli proteins in situ. Using enrichment approaches followed by LC-MS/MS, phosphosites were identified allowing motifs associated with auto- and transphosphorylation to be characterized. Four lines of evidence suggest that transphosphorylation of E. coli proteins by BRI1 is specific and therefore provides meaningful results: (1) phosphorylation is not correlated with bacterial protein abundance; (2) phosphosite stoichiometry, estimated by spectral counting, is also not related to protein abundance; (3) a transphosphorylation motif emerged with strong preference for basic residues both N- and C-terminal to the phosphosites; and (4) other protein kinases (BAK1, PEPR1, FLS2, and CDPKβ) phosphorylated a distinct set of E. coli proteins and phosphosites. The E. coli transphosphorylation assay can be applied broadly to protein kinases and provides a convenient and powerful system to elucidate kinase specificity.
Collapse
Affiliation(s)
- Xia Wu
- Department of Plant Biology, University of IllinoisUrbana, IL, USA
| | - Man-Ho Oh
- Department of Plant Biology, University of IllinoisUrbana, IL, USA
| | - Hyoung Seok Kim
- Department of Plant Biology, University of IllinoisUrbana, IL, USA
| | - Daniel Schwartz
- Department of Physiology and Neurobiology, University of ConnecticutStorrs, CT, USA
| | - Brian S. Imai
- Protein Sciences Facility, Carver Biotechnology Center, University of IllinoisUrbana, IL, USA
| | - Peter M. Yau
- Protein Sciences Facility, Carver Biotechnology Center, University of IllinoisUrbana, IL, USA
| | - Steven D. Clouse
- Department of Horticultural Science, North Carolina State UniversityRaleigh, NC, USA
| | - Steven C. Huber
- Department of Plant Biology, University of IllinoisUrbana, IL, USA
- Agricultural Research Service, United States Department of AgricultureUrbana, IL, USA
- *Correspondence: Steven C. Huber, Department of Plant Biology, University of Illinois, 1201 W. Gregory Drive, 197 ERML, Urbana, IL 61801-3838, USA. e-mail:
| |
Collapse
|
33
|
Technau M, Knispel M, Roth S. Molecular mechanisms of EGF signaling-dependent regulation of pipe, a gene crucial for dorsoventral axis formation in Drosophila. Dev Genes Evol 2011; 222:1-17. [PMID: 22198544 PMCID: PMC3291829 DOI: 10.1007/s00427-011-0384-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 11/29/2011] [Indexed: 01/28/2023]
Abstract
During Drosophila oogenesis the expression of the sulfotransferase Pipe in ventral follicle cells is crucial for dorsoventral axis formation. Pipe modifies proteins that are incorporated in the ventral eggshell and activate Toll signaling which in turn initiates embryonic dorsoventral patterning. Ventral pipe expression is the result of an oocyte-derived EGF signal which down-regulates pipe in dorsal follicle cells. The analysis of mutant follicle cell clones reveals that none of the transcription factors known to act downstream of EGF signaling in Drosophila is required or sufficient for pipe regulation. However, the pipe cis-regulatory region harbors a 31-bp element which is essential for pipe repression, and ovarian extracts contain a protein that binds this element. Thus, EGF signaling does not act by down-regulating an activator of pipe as previously suggested but rather by activating a repressor. Surprisingly, this repressor acts independent of the common co-repressors Groucho or CtBP.
Collapse
Affiliation(s)
- Martin Technau
- Institute for Developmental Biology, Biocenter, University of Cologne, Zuelpicher Straße 47b, 50674, Cologne, Germany
| | | | | |
Collapse
|
34
|
RBF and Rno promote photoreceptor differentiation onset through modulating EGFR signaling in the Drosophila developing eye. Dev Biol 2011; 359:190-8. [PMID: 21920355 DOI: 10.1016/j.ydbio.2011.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/21/2011] [Accepted: 08/28/2011] [Indexed: 01/13/2023]
Abstract
The retinoblastoma gene Rb is the prototype tumor suppressor and is conserved in Drosophila. We use the developing fly retina as a model system to investigate the role of Drosophila Rb (rbf) during differentiation. This report shows that mutation of rbf and rhinoceros (rno), which encodes a PHD domain protein, leads to a synergistic delay in photoreceptor cell differentiation in the developing eye disc. We show that this differentiation delay phenotype is caused by decreased levels of different components of the Epidermal Growth Factor Receptor (EGFR) signaling pathway in the absence of rbf and rno. We show that rbf is required for normal expression of Rhomboid proteins and activation of MAP kinase in the morphogenetic furrow (MF), while rno is required for the expression of Pointed (Pnt) and Ebi proteins, which are key factors that mediate EGFR signaling output in the nucleus. Interestingly, while removing the transcription activation function of dE2F1 is sufficient to suppress the synergistic differentiation delay, a mutant form of de2f1 that disrupts the binding with RBF but retains the transcription activation function does not mimic the effect of rbf loss. These observations suggest that RBF has additional functions besides dE2F1 binding that regulates EGFR signaling and photoreceptor differentiation.
Collapse
|
35
|
The lens in focus: a comparison of lens development in Drosophila and vertebrates. Mol Genet Genomics 2011; 286:189-213. [PMID: 21877135 DOI: 10.1007/s00438-011-0643-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/04/2011] [Indexed: 12/24/2022]
Abstract
The evolution of the eye has been a major subject of study dating back centuries. The advent of molecular genetics offered the surprising finding that morphologically distinct eyes rely on conserved regulatory gene networks for their formation. While many of these advances often stemmed from studies of the compound eye of the fruit fly, Drosophila melanogaster, and later translated to discoveries in vertebrate systems, studies on vertebrate lens development far outnumber those in Drosophila. This may be largely historical, since Spemann and Mangold's paradigm of tissue induction was discovered in the amphibian lens. Recent studies on lens development in Drosophila have begun to define molecular commonalities with the vertebrate lens. Here, we provide an overview of Drosophila lens development, discussing intrinsic and extrinsic factors controlling lens cell specification and differentiation. We then summarize key morphological and molecular events in vertebrate lens development, emphasizing regulatory factors and networks strongly associated with both systems. Finally, we provide a comparative analysis that highlights areas of research that would help further clarify the degree of conservation between the formation of dioptric systems in invertebrates and vertebrates.
Collapse
|
36
|
Tomlinson A, Mavromatakis YE, Struhl G. Three distinct roles for notch in Drosophila R7 photoreceptor specification. PLoS Biol 2011; 9:e1001132. [PMID: 21886484 PMCID: PMC3160325 DOI: 10.1371/journal.pbio.1001132] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/14/2011] [Indexed: 12/03/2022] Open
Abstract
During specification of the R7 photoreceptor in the <I>Drosophila</I> eye, activation of Notch signaling leads to multiple responses within the cell, including antagonistic ones. Receptor tyrosine kinases (RTKs) and Notch (N) proteins are different types of transmembrane receptors that transduce extracellular signals and control cell fate. Here we examine cell fate specification in the Drosophila retina and ask how N acts together with the RTKs Sevenless (Sev) and the EGF receptor (DER) to specify the R7 photoreceptor. The retina is composed of many hundred ommatidia, each of which grows by recruiting surrounding, undifferentiated cells and directing them to particular fates. The R7 photoreceptor derives from a cohort of three cells that are incorporated together following specification of the R2-R5 and R8 photoreceptors. Two cells of the cohort are specified as the R1/6 photoreceptor type by DER activation. These cells then activate N in the third cell (the R7 precursor). By manipulation of N and RTK signaling in diverse combinations we establish three roles for N in specifying the R7 fate. The first role is to impose a block to photoreceptor differentiation; a block that DER activation cannot overcome. The second role, paradoxically, is to negate the first; Notch activation up-regulates Sev expression, enabling the presumptive R7 cell to receive an RTK signal from R8 that can override the block. The third role is to specify the cell as an R7 rather than an R1/6 once RTK signaling has specified the cells as a photoreceptor. We speculate why N acts both to block and to facilitate photoreceptor differentiation, and provide a model for how N and RTK signaling act combinatorially to specify the R1/6 and R7 photoreceptors as well as the surrounding non-neuronal cone cells. Cells are often directed to their developmental fates by the signals they receive from other cells. The Drosophila eye has become a classic paradigm for studying such signaling, and in this system direct neighbor-to-neighbor signaling plays a large role. The R7 photoreceptor is directed to its fate by signals derived from two different neighboring cell types. One sends a signal that activates tyrosine kinase signaling in the R7 precursor, whereas the other activates the Notch signaling pathway. Here we examine Notch signaling and find that it induces three responses in the R7 precursor. We show that one role acts to inhibit the specification of the cell as a photoreceptor, while another role opposes this function, and acts to direct the cell to the photoreceptor fate. The third role specifies the cell as the specialized R7 photoreceptor rather than as the generic photoreceptor type. These results demonstrate that activation of a single signaling pathway can result in multiple cellular responses, even antagonistic ones.
Collapse
MESH Headings
- Animals
- Body Patterning
- Cell Differentiation
- Compound Eye, Arthropod/cytology
- Compound Eye, Arthropod/growth & development
- Compound Eye, Arthropod/metabolism
- Drosophila Proteins/genetics
- Drosophila Proteins/metabolism
- Drosophila melanogaster/growth & development
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Eye Proteins/genetics
- Eye Proteins/metabolism
- Gene Expression
- Gene Expression Regulation, Developmental
- Photoreceptor Cells, Invertebrate/cytology
- Photoreceptor Cells, Invertebrate/metabolism
- Photoreceptor Cells, Invertebrate/physiology
- Promoter Regions, Genetic
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Andrew Tomlinson
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York, United States of America.
| | | | | |
Collapse
|
37
|
Scortegagna M, Subtil T, Qi J, Kim H, Zhao W, Gu W, Kluger H, Ronai ZA. USP13 enzyme regulates Siah2 ligase stability and activity via noncatalytic ubiquitin-binding domains. J Biol Chem 2011; 286:27333-27341. [PMID: 21659512 PMCID: PMC3149327 DOI: 10.1074/jbc.m111.218214] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 05/23/2011] [Indexed: 02/04/2023] Open
Abstract
The RING finger E3 ubiquitin ligase Siah2 is implicated in control of diverse cellular biological events, including MAPK signaling and hypoxia. Here we demonstrate that Siah2 is subject to regulation by the deubiquitinating enzyme USP13. Overexpression of USP13 increases Siah2 stability by attenuating its autodegradation. Consequently, the ability of Siah2 to target its substrates prolyl hydroxylase 3 and Spry2 (Sprouty2) for ubiquitin-mediated proteasomal degradation is attenuated. Conversely, inhibition of USP13 expression with corresponding shRNA decreases the stability of both Siah2 and its substrate Spry2. Thus, USP13 limits Siah2 autodegradation and its ubiquitin ligase activity against its target substrates. Strikingly, the effect of USP13 on Siah2 is not mediated by its isopeptidase activity: mutations in its ubiquitin-binding sequences positioned within the ubiquitin-specific processing protease and ubiquitin-binding domains, but not within putative catalytic sites, abolish USP13 binding to and effect on Siah2 autodegradation and targeted ubiquitination. Notably, USP13 expression is attenuated in melanoma cells maintained under hypoxia, thereby relieving Siah2 inhibition and increasing its activity under low oxygen levels. Significantly, on melanoma tissue microarray, high nuclear expression of USP13 coincided with high nuclear expression of Siah2. Overall, this study identifies a new layer of Siah2 regulation mediated by USP13 binding to ubiquitinated Siah2 protein with a concomitant inhibitory effect on its activity under normoxia.
Collapse
Affiliation(s)
- Marzia Scortegagna
- From the Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92130
| | - Tony Subtil
- the Department of Medicine, Yale University, New Haven, Connecticut 06520, and
| | - Jianfei Qi
- From the Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92130
| | - Hyungsoo Kim
- From the Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92130
| | - Wenhui Zhao
- the Institute for Cancer Genetics, Herbert Irving Cancer Research Center, Columbia University, New York, New York 10032
| | - Wei Gu
- the Institute for Cancer Genetics, Herbert Irving Cancer Research Center, Columbia University, New York, New York 10032
| | - Harriet Kluger
- the Department of Medicine, Yale University, New Haven, Connecticut 06520, and
| | - Ze'ev A. Ronai
- From the Signal Transduction Program, Sanford-Burnham Medical Research Institute, La Jolla, California 92130
| |
Collapse
|
38
|
Herpes simplex virus immediate-early protein ICP0 is targeted by SIAH-1 for proteasomal degradation. J Virol 2011; 85:7644-57. [PMID: 21632771 DOI: 10.1128/jvi.02207-10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Herpes simplex virus (HSV) immediate-early protein ICP0 is a transcriptional activator with E3 ubiquitin ligase activity that induces the degradation of ND10 proteins, including the promyelocytic leukemia protein (PML) and Sp100. Moreover, ICP0 has a role in the derepression of viral genomes and in the modulation of the host interferon response to virus infection. Here, we report that ICP0 interacts with SIAH-1, a cellular E3 ubiquitin ligase that is involved in multiple cellular pathways and is itself capable of mediating PML degradation. This novel virus-host interaction profoundly stabilized SIAH-1 and recruited this cellular E3 ligase into ICP0-containing nuclear bodies. Moreover, SIAH-1 mediated the polyubiquitination of HSV ICP0 in vitro and in vivo. After infection of SIAH-1 knockdown cells with HSV, higher levels of ICP0 were produced, ICP0 was less ubiquitinated, and the half-life of this multifunctional viral regulatory protein was increased. These results indicate an inhibitory role of SIAH-1 during lytic infection by targeting ICP0 for proteasomal degradation.
Collapse
|
39
|
Sirover MA. On the functional diversity of glyceraldehyde-3-phosphate dehydrogenase: biochemical mechanisms and regulatory control. Biochim Biophys Acta Gen Subj 2011; 1810:741-51. [PMID: 21640161 DOI: 10.1016/j.bbagen.2011.05.010] [Citation(s) in RCA: 213] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND New studies provide evidence that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is not simply a classical glycolytic protein of little interest. Instead, it is a multifunctional protein with significant activity in a number of fundamental cell pathways. GAPDH is a highly conserved gene and protein, with a single mRNA transcribed from a unique gene. Control mechanisms must exist which regulate its functional diversity. SCOPE OF REVIEW This review focuses on new, timely studies defining not only its diverse activities but also those which define the regulatory mechanisms through which those functions may be controlled. The reader is referred to the author's prior review for the consideration of past reports which first indicated GAPDH multiple activities (Sirover, Biochim. Biophys. Acta 1432 (1999) 159-184.) CONCLUSIONS These investigations demonstrate fundamental roles of GAPDH in vivo, dynamic changes in its subcellular localization, and the importance of posttranslational modifications as well as protein:protein interactions as regulatory control mechanisms. GENERAL SIGNIFICANCE GAPDH is the prototype "moonlighting" protein which exhibits activities distinct from their classically identified functions. Their participation in diverse cell pathways is essential. Regulatory mechanisms exist which control those diverse activities as well as changes in their subcellular localization as a consequence of those new functions.
Collapse
Affiliation(s)
- Michael A Sirover
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| |
Collapse
|
40
|
Reddy BA, Bajpe PK, Bassett A, Moshkin YM, Kozhevnikova E, Bezstarosti K, Demmers JAA, Travers AA, Verrijzer CP. Drosophila transcription factor Tramtrack69 binds MEP1 to recruit the chromatin remodeler NuRD. Mol Cell Biol 2010; 30:5234-44. [PMID: 20733004 PMCID: PMC2953047 DOI: 10.1128/mcb.00266-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/14/2010] [Accepted: 07/18/2010] [Indexed: 11/20/2022] Open
Abstract
ATP-dependent chromatin-remodeling complexes (remodelers) are essential regulators of chromatin structure and gene transcription. How remodelers can act in a gene-selective manner has remained enigmatic. A yeast two-hybrid screen for proteins binding the Drosophila transcription factor Tramtrack69 (TTK69) identified MEP1. Proteomic characterization revealed that MEP1 is a tightly associated subunit of the NuRD remodeler, harboring the Mi2 enzymatic core ATPase. In addition, we identified the fly homolog of human Deleted in oral cancer 1 (DOC1), also known as CDK2-associated protein 1 (CDK2AP1), as a bona fide NuRD subunit. Biochemical and genetic assays supported the functional association between MEP1, Mi2, and TTK69. Genomewide expression analysis established that TTK69, MEP1, and Mi2 cooperate closely to control transcription. The TTK69 transcriptome profile correlates poorly with remodelers other than NuRD, emphasizing the selectivity of remodeler action. On the genes examined, TTK69 is able to bind chromatin in the absence of NuRD, but targeting of NuRD is dependent on TTK69. Thus, there appears to be a hierarchical relationship in which transcription factor binding precedes remodeler recruitment.
Collapse
Affiliation(s)
- B. Ashok Reddy
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Prashanth Kumar Bajpe
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Andrew Bassett
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Yuri M. Moshkin
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Elena Kozhevnikova
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Karel Bezstarosti
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Jeroen A. A. Demmers
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - Andrew A. Travers
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| | - C. Peter Verrijzer
- Department of Biochemistry and Centre for Biomedical Genetics, Proteomics Centre, Erasmus University Medical Centre, P.O. Box 1738, 3000 DR Rotterdam, Netherlands, MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB22QH, United Kingdom
| |
Collapse
|
41
|
Graham TGW, Tabei SMA, Dinner AR, Rebay I. Modeling bistable cell-fate choices in the Drosophila eye: qualitative and quantitative perspectives. Development 2010; 137:2265-78. [PMID: 20570936 PMCID: PMC2889600 DOI: 10.1242/dev.044826] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A major goal of developmental biology is to understand the molecular mechanisms whereby genetic signaling networks establish and maintain distinct cell types within multicellular organisms. Here, we review cell-fate decisions in the developing eye of Drosophila melanogaster and the experimental results that have revealed the topology of the underlying signaling circuitries. We then propose that switch-like network motifs based on positive feedback play a central role in cell-fate choice, and discuss how mathematical modeling can be used to understand and predict the bistable or multistable behavior of such networks.
Collapse
Affiliation(s)
- Thomas G. W. Graham
- Ben May Department for Cancer Research, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | - S. M. Ali Tabei
- James Franck Institute, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | - Aaron R. Dinner
- James Franck Institute, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| | - Ilaria Rebay
- Ben May Department for Cancer Research, University of Chicago, 929 East 57th Street, Chicago, IL 60637, USA
| |
Collapse
|
42
|
Park BS, Eo HJ, Jang IC, Kang HG, Song JT, Seo HS. Ubiquitination of LHY by SINAT5 regulates flowering time and is inhibited by DET1. Biochem Biophys Res Commun 2010; 398:242-6. [PMID: 20599732 DOI: 10.1016/j.bbrc.2010.06.067] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 06/16/2010] [Indexed: 12/21/2022]
Abstract
Ubiquitin is a small polypeptide and ubiquitination is the post-translational modification by ubiquitin protein, resulting in degradation of target proteins by the 26S proteasome complex. Here, we found that E3 ubiquitin ligase SINAT5, an Arabidopsis homologue of the Drosophila SINA RING-finger protein, interacts directly with LHY, a component of the circadian oscillator, and DET1, a negative regulator of light-regulated gene expression. We also found that SINAT5 has E3 ubiquitination activity for LHY but not for DET1. Interestingly, LHY ubiquitination by SINAT5 was inhibited by DET1. Late flowering of sinat5 mutants indicates that flowering time can be controlled by DET1 through regulation of LHY stability by SINAT5.
Collapse
Affiliation(s)
- Bong Soo Park
- Department of Plant Science, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 151-921, Republic of Korea
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
BTB-zinc finger transcription factors play many important roles in metazoan development. In these proteins, the BTB domain is critical for dimerization and for recruiting cofactors to target genes. Identification of these cofactors is important for understanding how BTB-zinc finger proteins influence transcription. Here we show that the novel but conserved protein EOR-2 is an obligate binding partner of the BTB-zinc finger protein EOR-1 in Caenorhabditis elegans. EOR-1 and EOR-2 function together to promote multiple Ras/ERK-dependent cell fates during development, and we show that EOR-1 is a robust substrate of ERK in vitro. A point mutation (L81F) in the EOR-1 BTB domain reduces both ERK phosphorylation and EOR-2 binding and eliminates all detectable biological function without affecting EOR-1 expression levels, localization, or dimerization. This point mutation lies near the predicted charged pocket region of the EOR-1 BTB dimer, a region that, in other BTB-zinc finger proteins, has been proposed to interact with corepressors or coactivators. We also show that a conserved zinc finger-like motif in EOR-2 is required for binding to EOR-1, that the interaction between EOR-1 and EOR-2 is direct, and that EOR-2 can bind to the human BTB-zinc finger protein PLZF. We propose that EOR-2 defines a new family of cofactors for BTB-zinc finger transcription factors that may have conserved roles in other organisms.
Collapse
Affiliation(s)
- Kelly Howell
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104 and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Swathi Arur
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104 and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Tim Schedl
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104 and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Meera V. Sundaram
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104 and Department of Genetics, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
44
|
Schneider G, Filipek A. S100A6 binding protein and Siah-1 interacting protein (CacyBP/SIP): spotlight on properties and cellular function. Amino Acids 2010; 41:773-80. [PMID: 20182755 DOI: 10.1007/s00726-010-0498-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 01/23/2010] [Indexed: 12/14/2022]
Abstract
The CacyBP/SIP protein (S100A6 binding protein and Siah-1 interacting protein) was originally discovered in Ehrlich ascites tumor cells as a S100A6 (calcyclin) target (Filipek and Wojda in Biochem J 320:585-587, 1996; Filipek and Kuźnicki in J Neurochem 70(5):1793-1798, 1998) and later on as a Siah-1 interacting protein (Matsuzawa and Reed in Mol Cell 7(5):915-926, 2001). CacyBP/SIP binds several target proteins such as some calcium binding proteins of the S100 family (Filipek et al. in J Biol Chem 277(32):28848-28852, 2002), Skp1 (Matsuzawa and Reed in Mol Cell 7(5):915-926, 2001), tubulin (Schneider et al. in Biochim Biophys Acta 1773(11):1628-1636, 2007) and ERK1/2 (Kilanczyk et al. in Biochem Biophys Res Commun 380:54-59, 2009). Studies concerning distribution of CacyBP/SIP show that it is present in various tissues and that a particularly high level of CacyBP/SIP is observed in brain (Jastrzebska et al. in J Histochem Cytochem 48(9):1195-1202, 2000). Regarding the function of CacyBP/SIP, there are some reports suggesting its role in cellular processes such as ubiquitination, proliferation, differentiation, tumorigenesis, cytoskeletal rearrangement or regulation of transcription. This review describes the properties of CacyBP/SIP and summarizes all findings concerning its cellular function.
Collapse
Affiliation(s)
- Gabriela Schneider
- Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093, Warsaw, Poland
| | | |
Collapse
|
45
|
Sobieszczuk DF, Poliakov A, Xu Q, Wilkinson DG. A feedback loop mediated by degradation of an inhibitor is required to initiate neuronal differentiation. Genes Dev 2010; 24:206-18. [PMID: 20080956 DOI: 10.1101/gad.554510] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neuronal differentiation is regulated by proneural genes that promote neurogenesis and inhibitory mechanisms that maintain progenitors. This raises the question of how the up-regulation of proneural genes required to initiate neurogenesis occurs in the presence of such inhibition. We carried out loss and gain of gene function, an interaction screen for binding partners, and biochemical analyses to uncover the regulation, developmental role, and mechanism of action of a ubiquitination adaptor protein, Btbd6a (BTB domain containing 6a). We find that the proneural gene neurog1 up-regulates btbd6a, which in turn is required for up-regulation of neurog1. Btbd6a is an adaptor for the Cul3 ubiquitin ligase complex, and we find that it binds to the transcriptional repressor Plzf (promyelocytic leukemia zinc finger). Btbd6a promotes the relocation of Plzf from nucleus to cytoplasm and targets Plzf for ubiquitination and degradation. plzfa is expressed widely in the neural epithelium; when overexpressed, it inhibits neurogenesis, and this inhibition is reversed by btbd6a. The antagonism of endogenous plzfa by btbd6a is required for neurogenesis, since the block in neuronal differentiation caused by btbd6a knockdown is alleviated by plzfa knockdown. These findings reveal a feedback loop mediated by degradation of an inhibitor that is essential for progenitors to undergo the transition to neuronal differentiation.
Collapse
Affiliation(s)
- Dorothy F Sobieszczuk
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, United Kingdom
| | | | | | | |
Collapse
|
46
|
Siddall NA, Hime GR, Pollock JA, Batterham P. Ttk69-dependent repression of lozenge prevents the ectopic development of R7 cells in the Drosophila larval eye disc. BMC DEVELOPMENTAL BIOLOGY 2009; 9:64. [PMID: 20003234 PMCID: PMC2797499 DOI: 10.1186/1471-213x-9-64] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 12/09/2009] [Indexed: 11/18/2022]
Abstract
Background During the development of the Drosophila eye, specific cell types differentiate from an initially equipotent group of uncommitted precursor cells. The lozenge (lz) gene, which is a member of the Runt family of transcriptional regulators, plays a pivotal role in mediating this process through regulating the expression of several fate-specifying transcription factors. However, the regulation of lz, and the control of lz expression levels in different cell types is not fully understood. Results Here, we show a genetic interaction between Tramtrack69 (Ttk69) a key transcriptional repressor and an inhibitor of neuronal fate specification, and lz, the master patterning gene of cells posterior to the morphogenetic furrow in the Drosophila eye disc. Loss of Ttk69 expression causes the development of ectopic R7 cells in the third instar eye disc, with these cells being dependent upon Lz for their development. Using the binary UAS Gal4 system, we show that overexpression of Ttk69 causes the loss of lz-dependent differentiating cells, and a down-regulation of Lz expression in the developing eye. The loss of lz-dependent cells can be rescued by overexpressing lz via a GMR-lz transgene. We provide additional data showing that factors functioning upstream of Ttk69 in eye development regulate lz in a Ttk69-dependent manner. Conclusions Our results lead us to conclude that Ttk69 can either directly or indirectly repress lz gene expression to prevent the premature development of R7 precursor cells in the developing eye of Drosophila. We therefore define a mechanism for the tight regulatory control of the master pre-patterning gene, lz, in early Drosophila eye development and provide insight into how differential levels of lz expression can be achieved to effect specific cell fate outcomes.
Collapse
Affiliation(s)
- Nicole A Siddall
- Department of Genetics, University of Melbourne, Parkville, Vic 3010, Australia.
| | | | | | | |
Collapse
|
47
|
Nakayama K. Cellular signal transduction of the hypoxia response. J Biochem 2009; 146:757-65. [PMID: 19864435 DOI: 10.1093/jb/mvp167] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Cells induce the hypoxia responses to adapt to the environment when organisms are exposed to a low oxygen environment. The hypoxia response leads to the activation of multiple cellular signalling pathways involved in regulation of respiration, metabolism, cell survival and so forth. Hypoxia-Inducible-Factor (HIF) pathway plays a central role during the hypoxia response as its expression and activity are regulated in an oxygen-dependent manner and it also regulates the expression of multiple hypoxia responsive genes. The expression of HIF is regulated by proline hydroxylation, which is mediated by HIF prolyl-hydroxylase named PHD. The hydroxylated HIF-alpha subunit is degraded via the ubiquitin-proteasome pathway. The PHD activity needs to be strictly regulated to ensure the stabilization of HIF under hypoxic conditions, because PHD leads to HIF degradation. This review describes the regulatory mechanism of HIF stability and activity under normoxia and hypoxic conditions. Furthermore, the role of the HIF-independent pathways during the hypoxia response, which is as important as the HIF pathway, will also be described.
Collapse
Affiliation(s)
- Koh Nakayama
- Medical Top Track program, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
48
|
Twomey E, Li Y, Lei J, Sodja C, Ribecco-Lutkiewicz M, Smith B, Fang H, Bani-Yaghoub M, McKinnell I, Sikorska M. Regulation of MYPT1 stability by the E3 ubiquitin ligase SIAH2. Exp Cell Res 2009; 316:68-77. [PMID: 19744480 DOI: 10.1016/j.yexcr.2009.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 08/25/2009] [Accepted: 09/01/2009] [Indexed: 10/20/2022]
Abstract
Myosin phosphatase target subunit 1 (MYPT1), together with catalytic subunit of type1 delta isoform (PP1cdelta) and a small 20-kDa regulatory unit (M20), form a heterotrimeric holoenzyme, myosin phosphatase (MP), which is responsible for regulating the extent of myosin light chain phosphorylation. Here we report the identification and characterization of a molecular interaction between Seven in absentia homolog 2 (SIAH2) and MYPT1 that resulted in the proteasomal degradation of the latter in mammalian cells, including neurons and glia. The interaction involved the substrate binding domain of SIAH2 (aa 116-324) and a central region of MYPT1 (aa 445-632) containing a degenerate consensus Siah-binding motif RLAYVAP (aa 493-499) evolutionally conserved from fish to humans. These findings suggest a novel mechanism whereby the ability of MP to modulate myosin light chain might be regulated by the degradation of its targeting subunit MYPT1 through the SIAH2-ubiquitin-proteasomal pathway. In this manner, the turnover of MYPT1 would serve to limit the duration and/or magnitude of MP activity required to achieve a desired physiological effect.
Collapse
Affiliation(s)
- Erin Twomey
- Neurogenesis and Brain Repair Group, Institute for Biological Sciences, National Research Council of Canada, 1200 Montreal Road, M-54, Ottawa, Canada ON K1A 0R6
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Shi Y, Noll M. Determination of cell fates in the R7 equivalence group of the Drosophila eye by the concerted regulation of D-Pax2 and TTK88. Dev Biol 2009; 331:68-77. [PMID: 19406115 DOI: 10.1016/j.ydbio.2009.04.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 04/06/2009] [Accepted: 04/22/2009] [Indexed: 11/30/2022]
Abstract
In the developing Drosophila eye, the precursors of the neuronal photoreceptor cells R1/R6/R7 and non-neuronal cone cells share the same developmental potential and constitute the R7 equivalence group. It is not clear how cells of this group elaborate their distinct fates. Here we show that both TTK88 and D-Pax2 play decisive roles in cone cell development and act in concert to transform developing R1/R6/R7 into cone cells: while TTK88 blocks neuronal development, D-Pax2 promotes cone cell specification. In addition, ectopic TTK88 in R cells induces apoptosis, which is suppressed by ectopic D-Pax2. We further demonstrate that Phyllopod (Phyl), previously shown to promote the neuronal fate in R1/R6/R7 by targeting TTK for degradation, also inhibits D-Pax2 transcription to prevent cone cell specification. Thus, the fates of R1/R6/R7 and cone cells are determined by a dual mechanism that coordinately activates one fate while inhibiting the other.
Collapse
Affiliation(s)
- Yandong Shi
- Institute for Molecular Biology, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | |
Collapse
|
50
|
Li X, Cassidy JJ, Reinke CA, Fischboeck S, Carthew RW. A microRNA imparts robustness against environmental fluctuation during development. Cell 2009; 137:273-82. [PMID: 19379693 PMCID: PMC2674871 DOI: 10.1016/j.cell.2009.01.058] [Citation(s) in RCA: 368] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 11/25/2008] [Accepted: 01/29/2009] [Indexed: 11/20/2022]
Abstract
The microRNA miR-7 is perfectly conserved from annelids to humans, and yet some of the genes that it regulates in Drosophila are not regulated in mammals. We have explored the role of lineage restricted targets, using Drosophila, in order to better understand the evolutionary significance of microRNA-target relationships. From studies of two well characterized developmental regulatory networks, we find that miR-7 functions in several interlocking feedback and feedforward loops, and propose that its role in these networks is to buffer them against perturbation. To directly demonstrate this function for miR-7, we subjected the networks to temperature fluctuation and found that miR-7 is essential for the maintenance of regulatory stability under conditions of environmental flux. We suggest that some conserved microRNAs like miR-7 may enter into novel genetic relationships to buffer developmental programs against variation and impart robustness to diverse regulatory networks.
Collapse
Affiliation(s)
| | | | - Catherine A. Reinke
- Department of Biochemistry, Molecular Biology and Cell Biology, 2205 Tech Drive, Northwestern University, Evanston, Illinois 60208, USA
| | - Stephen Fischboeck
- Department of Biochemistry, Molecular Biology and Cell Biology, 2205 Tech Drive, Northwestern University, Evanston, Illinois 60208, USA
| | - Richard W. Carthew
- Department of Biochemistry, Molecular Biology and Cell Biology, 2205 Tech Drive, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|