1
|
Wang G, Shen G, Xu C, Guo Y, Zhang W, Wang Q, Zhu Y. Caspase-8 promotes innate immunity in the Chinese mitten crab by regulating the expression of antimicrobial peptides and apoptosis in hemocyte. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105308. [PMID: 39724998 DOI: 10.1016/j.dci.2024.105308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
In mammals, caspase-8 primarily functions as an initiator caspase that regulates apoptosis, while in Drosophila, the caspase-8 ortholog DREDD not only induces apoptosis during development but also regulates antimicrobial peptides (AMPs) expression during Gram-negative bacterial infection-induced immune responses. However, the immune-related function of caspase-8 in the crustacean remains unknown. In the present study, the open reading frame of EsCaspase-8 was cloned from the Chinese mitten crab (Eriocheir sinensis). The deduced EsCaspase-8 protein sequence contained only one death effector domain (DED) and a cysteine aspartase cysteine structural domain. The EsCaspase-8 expression was significantly induced after 6 h of Vibrio parahaemolyticus infection and continued to 24 h in hemocyte. Knocking down EsCaspase-8 expression in hemocytes significantly inhibited Relish's nuclear translocation and suppressed the expression of AMPs, including Crustin 1, Crustin 2, Lysosome, and double WAP domain, after V. parahaemolyticus infection. Furthermore, the knockdown of EsCaspase-8 in vivo significantly inhibited hemocyte apoptosis post-bacterial infection. These results demonstrated that EsCaspase-8 can promote antibacterial activities by regulating the expression of AMPs through activation of Relish nuclear translocation in Chinese mitten crabs, thus acting as a critical positive regulator in innate immunity. In addition, EsCaspase-8 also has the function of inducing hemocyte apoptosis. These findings expand our understanding of the molecular mechanisms underlying crustacean immune responses and provide a foundation for future research to improve disease resistance.
Collapse
Affiliation(s)
- Guangyu Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Guoqing Shen
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Chaohui Xu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yanan Guo
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qun Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Youting Zhu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
2
|
Szlanka T, Lukacsovich T, Bálint É, Virágh E, Szabó K, Hajdu I, Molnár E, Lin YH, Zvara Á, Kelemen-Valkony I, Méhi O, Török I, Hegedűs Z, Kiss B, Ramasz B, Magdalena LM, Puskás L, Mechler BM, Fónagy A, Asztalos Z, Steinbach G, Žurovec M, Boros I, Kiss I. Dominant suppressor genes of p53-induced apoptosis in Drosophila melanogaster. G3 (BETHESDA, MD.) 2024; 14:jkae149. [PMID: 38985658 PMCID: PMC11373661 DOI: 10.1093/g3journal/jkae149] [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: 02/06/2024] [Revised: 02/06/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024]
Abstract
One of the major functions of programmed cell death (apoptosis) is the removal of cells that suffered oncogenic mutations, thereby preventing cancerous transformation. By making use of a Double-Headed-EP (DEP) transposon, a P element derivative made in our laboratory, we made an insertional mutagenesis screen in Drosophila melanogaster to identify genes that, when overexpressed, suppress the p53-activated apoptosis. The DEP element has Gal4-activatable, outward-directed UAS promoters at both ends, which can be deleted separately in vivo. In the DEP insertion mutants, we used the GMR-Gal4 driver to induce transcription from both UAS promoters and tested the suppression effect on the apoptotic rough eye phenotype generated by an activated UAS-p53 transgene. By DEP insertions, 7 genes were identified, which suppressed the p53-induced apoptosis. In 4 mutants, the suppression effect resulted from single genes activated by 1 UAS promoter (Pka-R2, Rga, crol, and Spt5). In the other 3 (Orct2, Polr2M, and stg), deleting either UAS promoter eliminated the suppression effect. In qPCR experiments, we found that the genes in the vicinity of the DEP insertion also showed an elevated expression level. This suggested an additive effect of the nearby genes on suppressing apoptosis. In the eukaryotic genomes, there are coexpressed gene clusters. Three of the DEP insertion mutants are included, and 2 are in close vicinity of separate coexpressed gene clusters. This raises the possibility that the activity of some of the genes in these clusters may help the suppression of the apoptotic cell death.
Collapse
Affiliation(s)
- Tamás Szlanka
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Tamás Lukacsovich
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Éva Bálint
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Erika Virágh
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Kornélia Szabó
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Ildikó Hajdu
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Enikő Molnár
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Yu-Hsien Lin
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Ildikó Kelemen-Valkony
- Cellular Imaging Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Orsolya Méhi
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - István Török
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Zoltán Hegedűs
- Bioinformatics Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Brigitta Kiss
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Beáta Ramasz
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Laura M Magdalena
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - László Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Bernard M Mechler
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Adrien Fónagy
- Centre for Agricultural Sciences, Plant Protection Institute, 1022 Budapest, Hungary
| | - Zoltán Asztalos
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Aktogen Hungary Ltd., 6726 Szeged, Hungary
| | - Gábor Steinbach
- Cellular Imaging Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Michal Žurovec
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
| | - Imre Boros
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - István Kiss
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| |
Collapse
|
3
|
Cumming T, Levayer R. Toward a predictive understanding of epithelial cell death. Semin Cell Dev Biol 2024; 156:44-57. [PMID: 37400292 DOI: 10.1016/j.semcdb.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Epithelial cell death is highly prevalent during development and tissue homeostasis. While we have a rather good understanding of the molecular regulators of programmed cell death, especially for apoptosis, we still fail to predict when, where, how many and which specific cells will die in a tissue. This likely relies on the much more complex picture of apoptosis regulation in a tissular and epithelial context, which entails cell autonomous but also non-cell autonomous factors, diverse feedback and multiple layers of regulation of the commitment to apoptosis. In this review, we illustrate this complexity of epithelial apoptosis regulation by describing these different layers of control, all demonstrating that local cell death probability is a complex emerging feature. We first focus on non-cell autonomous factors that can locally modulate the rate of cell death, including cell competition, mechanical input and geometry as well as systemic effects. We then describe the multiple feedback mechanisms generated by cell death itself. We also outline the multiple layers of regulation of epithelial cell death, including the coordination of extrusion and regulation occurring downstream of effector caspases. Eventually, we propose a roadmap to reach a more predictive understanding of cell death regulation in an epithelial context.
Collapse
Affiliation(s)
- Tom Cumming
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France; Sorbonne Université, Collège Doctoral, F75005 Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
4
|
Cui Q, Huang C, Liu JY, Zhang JT. Small Molecule Inhibitors Targeting the "Undruggable" Survivin: The Past, Present, and Future from a Medicinal Chemist's Perspective. J Med Chem 2023; 66:16515-16545. [PMID: 38092421 PMCID: PMC11588358 DOI: 10.1021/acs.jmedchem.3c01130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Survivin, a homodimeric protein and a member of the IAP family, plays a vital function in cell survival and cycle progression by interacting with various proteins and complexes. Its expression is upregulated in cancers but not detectable in normal tissues. Thus, it has been regarded and validated as an ideal cancer target. However, survivin is "undruggable" due to its lack of enzymatic activities or active sites for small molecules to bind/inhibit. Academic and industrial laboratories have explored different strategies to overcome this hurdle over the past two decades, with some compounds advanced into clinical testing. These strategies include inhibiting survivin expression, its interaction with binding partners and homodimerization. Here, we provide comprehensive analyses of these strategies and perspective on different small molecule survivin inhibitors to help drug discovery targeting "undruggable" proteins in general and survivin specifically with a true survivin inhibitor that will prevail in the foreseeable future.
Collapse
Affiliation(s)
- Qingbin Cui
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
5
|
Lu MX, He FJ, Zhu F, Du YZ. The regulation of inhibitor of apoptosis proteins (IAPs) during the apoptosis of Cotesia chilonis. Front Physiol 2023; 14:1328167. [PMID: 38192740 PMCID: PMC10773855 DOI: 10.3389/fphys.2023.1328167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Inhibitor of apoptosis proteins (IAPs) are crucial components of apoptosis that perform vital roles in the regulation of caspase activity in organisms. In this study, two IAPs genes were identified from Cotesia chilonis, the dominant parasitic wasp of Chilo suppressalis. CcIAP1 gene is a typical IAP and contains two BIR domains and a RING domain, whereas CcIAP gene is an atypical IAP1 only containing two BIR domains. Phylogenetic analysis indicated that CcIAP1 and CcIAP were grouped with other Hymenopteran IAPs and IAP1 in C. suppressalis. Real-time quantitative PCR revealed that CcIAP1 and CcIAP genes were both highly induced at -6°C and 30°C, and expression was highest at the third instar stage. The expression of CcIAP1 and CcIAP genes were significantly induced during parasitism of C. suppressalis, and the 7-d time point resulted in the highest expression levels for both genes, in which was an advanced stage of larval development of C. chilonis. RNAi experiments showed that CcIAP1 gene was the key IAP in the regulation of apoptosis of C. chilonis and its host. In conclusion, CcIAP1 and CcIAP correlate with the development of C. chilonis and their responses to temperature stress.
Collapse
Affiliation(s)
- Ming-Xing Lu
- College of Plant Protection and Institute of Applied Entomology, Yangzhou University, Yangzhou, China
| | - Fu-Jing He
- Plant Protection and Quarantine Station of Jiangsu Province, Nanjing, China
| | - Feng Zhu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University, Yangzhou, China
| | - Yu-Zhou Du
- College of Plant Protection and Institute of Applied Entomology, Yangzhou University, Yangzhou, China
- Wuxi Vocational Institute of Commerce, Wuxi, China
| |
Collapse
|
6
|
Zheng J, Guo Y, Shi C, Yang S, Xu W, Ma X. Differential Ire1 determines loser cell fate in tumor-suppressive cell competition. Cell Rep 2023; 42:113303. [PMID: 37924514 DOI: 10.1016/j.celrep.2023.113303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/31/2023] [Accepted: 10/03/2023] [Indexed: 11/06/2023] Open
Abstract
Tumor-suppressive cell competition (TSCC) is a conserved surveillance mechanism in which neighboring cells actively eliminate oncogenic cells. Despite overwhelming studies showing that the unfolded protein response (UPR) is dysregulated in various tumors, it remains debatable whether the UPR restrains or promotes tumorigenesis. Here, using Drosophila eye epithelium as a model, we uncover a surprising decisive role of the Ire1 branch of the UPR in regulating cell polarity gene scribble (scrib) loss-induced TSCC. Both mutation and hyperactivation of Ire1 accelerate elimination of scrib clones via inducing apoptosis and autophagy, respectively. Unexpectedly, relative Ire1 activity is also crucial for determining loser cell fate, as dysregulating Ire1 signaling in the surrounding healthy cells reversed the "loser" status of scrib clones by decreasing their apoptosis. Furthermore, we show that Ire1 is required for cell competition in mammalian cells. Together, these findings provide molecular insights into scrib-mediated TSCC and highlight Ire1 as a key determinant of loser cell fate.
Collapse
Affiliation(s)
- Jiadong Zheng
- Fudan University, Shanghai 200433, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Yifan Guo
- Fudan University, Shanghai 200433, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Changyi Shi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Shuai Yang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Wenyan Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China.
| | - Xianjue Ma
- Fudan University, Shanghai 200433, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
7
|
Heron R, Amato C, Wood W, Davidson AJ. Understanding the diversity and dynamics of in vivo efferocytosis: Insights from the fly embryo. Immunol Rev 2023; 319:27-44. [PMID: 37589239 PMCID: PMC10952863 DOI: 10.1111/imr.13266] [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: 04/03/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
The clearance of dead and dying cells, termed efferocytosis, is a rapid and efficient process and one that is critical for organismal health. The extraordinary speed and efficiency with which dead cells are detected and engulfed by immune cells within tissues presents a challenge to researchers who wish to unravel this fascinating process, since these fleeting moments of uptake are almost impossible to catch in vivo. In recent years, the fruit fly (Drosophila melanogaster) embryo has emerged as a powerful model to circumvent this problem. With its abundance of dying cells, specialist phagocytes and relative ease of live imaging, the humble fly embryo provides a unique opportunity to catch and study the moment of cell engulfment in real-time within a living animal. In this review, we explore the recent advances that have come from studies in the fly, and how live imaging and genetics have revealed a previously unappreciated level of diversity in the efferocytic program. A variety of efferocytic strategies across the phagocytic cell population ensure efficient and rapid clearance of corpses wherever death is encountered within the varied and complex setting of a multicellular living organism.
Collapse
Affiliation(s)
- Rosalind Heron
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Clelia Amato
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Will Wood
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Andrew J. Davidson
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
- School of Cancer SciencesWolfson Wohl Cancer Research Centre, University of GlasgowGlasgowUK
| |
Collapse
|
8
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
9
|
Messer CL, McDonald JA. Rap1 promotes epithelial integrity and cell viability in a growing tissue. Dev Biol 2023; 501:1-19. [PMID: 37269969 DOI: 10.1016/j.ydbio.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 05/10/2023] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
Having intact epithelial tissues is critical for embryonic development and adult homeostasis. How epithelia respond to damaging insults or tissue growth while still maintaining intercellular connections and barrier integrity during development is poorly understood. The conserved small GTPase Rap1 is critical for establishing cell polarity and regulating cadherin-catenin cell junctions. Here, we identified a new role for Rap1 in maintaining epithelial integrity and tissue shape during Drosophila oogenesis. Loss of Rap1 activity disrupted the follicle cell epithelium and the shape of egg chambers during a period of major growth. Rap1 was required for proper E-Cadherin localization in the anterior epithelium and for epithelial cell survival. Both Myo-II and the adherens junction-cytoskeletal linker protein α-Catenin were required for normal egg chamber shape but did not strongly affect cell viability. Blocking the apoptotic cascade failed to rescue the cell shape defects caused by Rap1 inhibition. One consequence of increased cell death caused by Rap1 inhibition was the loss of polar cells and other follicle cells, which later in development led to fewer cells forming a migrating border cell cluster. Our results thus indicate dual roles for Rap1 in maintaining epithelia and cell survival in a growing tissue during development.
Collapse
Affiliation(s)
- C Luke Messer
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Jocelyn A McDonald
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
10
|
Spitzer DC, Sun WY, Rodríguez-Vargas A, Hariharan IK. The cell adhesion molecule Echinoid promotes tissue survival and separately restricts tissue overgrowth in Drosophila imaginal discs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552072. [PMID: 37577631 PMCID: PMC10418178 DOI: 10.1101/2023.08.04.552072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The interactions that cells in Drosophila imaginal discs have with their neighbors are known to regulate their ability to survive. In a screen of genes encoding cell surface proteins for gene knockdowns that affect the size or shape of mutant clones, we found that clones of cells with reduced levels of echinoid (ed) are fewer, smaller, and can be eliminated during development. In contrast, discs composed mostly of ed mutant tissue are overgrown. We find that ed mutant tissue has lower levels of the anti-apoptotic protein Diap1 and has increased levels of apoptosis which is consistent with the observed underrepresentation of ed mutant clones and the slow growth of ed mutant tissue. The eventual overgrowth of ed mutant tissue results not from accelerated growth, but from prolonged growth resulting from a failure to arrest growth at the appropriate final size. Ed has previously been shown to physically interact with multiple Hippo-pathway components and it has been proposed to promote Hippo pathway signaling, to exclude Yorkie (Yki) from the nucleus, and restrain the expression of Yki-target genes. We did not observe changes in Yki localization in ed mutant tissue and found decreased levels of expression of several Yorkie-target genes, findings inconsistent with the proposed effect of Ed on Yki. We did, however, observe increased expression of several Yki-target genes in wild-type cells neighboring ed mutant cells, which may contribute to elimination of ed mutant clones. Thus, ed has two distinct functions: an anti-apoptotic function by maintaining Diap1 levels, and a function to arrest growth at the appropriate final size. Both of these are unlikely to be explained by a simple effect on the Hippo pathway.
Collapse
Affiliation(s)
- Danielle C. Spitzer
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - William Y. Sun
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Anthony Rodríguez-Vargas
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| | - Iswar K. Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley CA 94720-3200
| |
Collapse
|
11
|
Factors Affecting Arbovirus Midgut Escape in Mosquitoes. Pathogens 2023; 12:pathogens12020220. [PMID: 36839492 PMCID: PMC9963182 DOI: 10.3390/pathogens12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Arboviral diseases spread by mosquitoes cause significant morbidity and mortality throughout much of the world. The treatment and prevention of these diseases through medication and vaccination is often limited, which makes controlling arboviruses at the level of the vector ideal. One way to prevent the spread of an arbovirus would be to stop its vector from developing a disseminated infection, which is required for the virus to make its way to the saliva of the mosquito to be potentially transmitted to a new host. The midgut of the mosquito provides one such opportunity to stop an arbovirus in its tracks. It has been known for many years that in certain arbovirus-vector combinations, or under certain circumstances, an arbovirus can infect and replicate in the midgut but is unable to escape from the tissue to cause disseminated infection. This situation is known as a midgut escape barrier. If we better understand why this barrier occurs, it might aid in the development of more informed control strategies. In this review, we discuss how the midgut escape barrier contributes to virus-vector specificity and possible mechanisms that may allow this barrier to be overcome in successful virus-vector combinations. We also discuss several of the known factors that either increase or decrease the likelihood of midgut escape.
Collapse
|
12
|
Nukala KM, Lilienthal AJ, Lye SH, Bassuk AG, Chtarbanova S, Manak JR. Downregulation of oxidative stress-mediated glial innate immune response suppresses seizures in a fly epilepsy model. Cell Rep 2023; 42:112004. [PMID: 36641750 PMCID: PMC9942582 DOI: 10.1016/j.celrep.2023.112004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/30/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Previous work in our laboratory has shown that mutations in prickle (pk) cause myoclonic-like seizures and ataxia in Drosophila, similar to what is observed in humans carrying mutations in orthologous PRICKLE genes. Here, we show that pk mutant brains show elevated, sustained neuronal cell death that correlates with increasing seizure penetrance, as well as an upregulation of mitochondrial oxidative stress and innate immune response (IIR) genes. Moreover, flies exhibiting more robust seizures show increased levels of IIR-associated target gene expression suggesting they may be linked. Genetic knockdown in glia of either arm of the IIR (Immune Deficiency [Imd] or Toll) leads to a reduction in neuronal death, which in turn suppresses seizure activity, with oxidative stress acting upstream of IIR. These data provide direct genetic evidence that oxidative stress in combination with glial-mediated IIR leads to progression of an epilepsy disorder.
Collapse
Affiliation(s)
- Krishna M Nukala
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Shu Hui Lye
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA; The Iowa Neuroscience Institute, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - J Robert Manak
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA; Department of Pediatrics, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
13
|
Ohta T, Tanimura T, Kimura KI. A gain-of-function mutation in head involution defective , Wrinkled, causes precocious cell death of wing epidermal cells in Drosophila. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000703. [PMID: 36606079 PMCID: PMC9808536 DOI: 10.17912/micropub.biology.000703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 01/07/2023]
Abstract
In Drosophila , wing epidermal cells undergo programmed cell death as the last step of metamorphosis. The aim of this study was to evaluate the role of hid , particularly the Wrinkled mutation ( hid W ), an allele of hid , in the cell death. The wing epithelial cell death is suppressed by loss-of-function mutation of hid , indicating that the death is governed by a cascade involving hid . Examination of the cell death in hid W showed that precocious death started at G stage, 3 h before eclosion. Thus, mutated-HID in the hid W mutant was activated at G stage, supporting the gain-of-function effect of hid W mutation.
Collapse
Affiliation(s)
- Takumi Ohta
- Laboratory of Biology, Hokkaido University of Education, Sapporo Campus, Sapporo, Japan
,
Department of Biology, Graduate School of Science, Kyushu University, Fukuoka, Japan
| | - Teiichi Tanimura
- Department of Biology, Graduate School of Science, Kyushu University, Fukuoka, Japan
,
Present address: Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Ken-ichi Kimura
- Laboratory of Biology, Hokkaido University of Education, Sapporo Campus, Sapporo, Japan
,
Correspondence to: Ken-ichi Kimura (
)
| |
Collapse
|
14
|
Wang Y, Lee H, Fear JM, Berger I, Oliver B, Przytycka TM. NetREX-CF integrates incomplete transcription factor data with gene expression to reconstruct gene regulatory networks. Commun Biol 2022; 5:1282. [PMID: 36418514 PMCID: PMC9684490 DOI: 10.1038/s42003-022-04226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
The inference of Gene Regulatory Networks (GRNs) is one of the key challenges in systems biology. Leading algorithms utilize, in addition to gene expression, prior knowledge such as Transcription Factor (TF) DNA binding motifs or results of TF binding experiments. However, such prior knowledge is typically incomplete, therefore, integrating it with gene expression to infer GRNs remains difficult. To address this challenge, we introduce NetREX-CF-Regulatory Network Reconstruction using EXpression and Collaborative Filtering-a GRN reconstruction approach that brings together Collaborative Filtering to address the incompleteness of the prior knowledge and a biologically justified model of gene expression (sparse Network Component Analysis based model). We validated the NetREX-CF using Yeast data and then used it to construct the GRN for Drosophila Schneider 2 (S2) cells. To corroborate the GRN, we performed a large-scale RNA-Seq analysis followed by a high-throughput RNAi treatment against all 465 expressed TFs in the cell line. Our knockdown result has not only extensively validated the GRN we built, but also provides a benchmark that our community can use for evaluating GRNs. Finally, we demonstrate that NetREX-CF can infer GRNs using single-cell RNA-Seq, and outperforms other methods, by using previously published human data.
Collapse
Affiliation(s)
- Yijie Wang
- Computer Science Department, Indiana University, Bloomington, IN, 47408, USA.
| | - Hangnoh Lee
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Justin M Fear
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Isabelle Berger
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA
| | - Brian Oliver
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, 50 South Drive, Bethesda, MD, 20892, USA.
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD, 20894, USA.
| |
Collapse
|
15
|
Gebreegziabher Amare M, Westrick NM, Keller NP, Kabbage M. The conservation of IAP-like proteins in fungi, and their potential role in fungal programmed cell death. Fungal Genet Biol 2022; 162:103730. [PMID: 35998750 DOI: 10.1016/j.fgb.2022.103730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
Programmed cell death (PCD) is a tightly regulated process which is required for survival and proper development of all cellular life. Despite this ubiquity, the precise molecular underpinnings of PCD have been primarily characterized in animals. Attempts to expand our understanding of this process in fungi have proven difficult as core regulators of animal PCD are apparently absent in fungal genomes, with the notable exception of a class of proteins referred to as inhibitors of apoptosis proteins (IAPs). These proteins are characterized by the conservation of a distinct Baculovirus IAP Repeat (BIR) domain and animal IAPs are known to regulate a number of processes, including cellular death, development, organogenesis, immune system maturation, host-pathogen interactions and more. IAP homologs are broadly conserved throughout the fungal kingdom, but our understanding of both their mechanism and role in fungal development/virulence is still unclear. In this review, we provide a broad and comparative overview of IAP function across taxa, with a particular focus on fungal processes regulated by IAPs. Furthermore, their putative modes of action in the absence of canonical interactors will be discussed.
Collapse
Affiliation(s)
| | - Nathaniel M Westrick
- Department of Plant Pathology, University of Wisconsin - Madison, Madison, WI, USA
| | - Nancy P Keller
- Department of Plant Pathology, University of Wisconsin - Madison, Madison, WI, USA
| | - Mehdi Kabbage
- Department of Plant Pathology, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Fasciclin 2 engages EGFR in an auto-stimulatory loop to promote imaginal disc cell proliferation in Drosophila. PLoS Genet 2022; 18:e1010224. [PMID: 35666718 PMCID: PMC9203005 DOI: 10.1371/journal.pgen.1010224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/16/2022] [Accepted: 04/28/2022] [Indexed: 12/04/2022] Open
Abstract
How cell to cell interactions control local tissue growth to attain a species-specific organ size is a central question in developmental biology. The Drosophila Neural Cell Adhesion Molecule, Fasciclin 2, is expressed during the development of neural and epithelial organs. Fasciclin 2 is a homophilic-interaction protein that shows moderate levels of expression in the proliferating epithelia and high levels in the differentiating non-proliferative cells of imaginal discs. Genetic interactions and mosaic analyses reveal a cell autonomous requirement of Fasciclin 2 to promote cell proliferation in imaginal discs. This function is mediated by the EGFR, and indirectly involves the JNK and Hippo signaling pathways. We further show that Fasciclin 2 physically interacts with EGFR and that, in turn, EGFR activity promotes the cell autonomous expression of Fasciclin 2 during imaginal disc growth. We propose that this auto-stimulatory loop between EGFR and Fasciclin 2 is at the core of a cell to cell interaction mechanism that controls the amount of intercalary growth in imaginal discs. A key problem in developmental biology is how species-specific organ size is determined. Control of organ growth occurs at different levels of organization, from the systemic to the cell to cell interaction level. During nervous system development cell contact interactions regulate axon growth. Here, we show that one of the cell adhesion molecules involved in controlling axon growth, the Drosophila NCAM ortholog Fasciclin 2, also controls epithelial organ growth and size. Fasciclin 2 is expressed in highly dynamic but moderate levels during cell proliferation in imaginal discs (precursor epithelial organs of the adult epidermis), and at much higher level in pre-differentiating and differentiating cells in imaginal discs. During imaginal disc growth cell interactions mediated by Fasciclin 2 promote Epidermal Growth Factor Receptor function and cell proliferation. In turn, Epidermal Growth Factor Receptor activity promotes Fasciclin 2 expression, creating a cell autonomous auto-stimulatory loop that maintains cell proliferation. This function of Fasciclin 2 is reciprocal to its reported function in pre-differentiating and differentiating cells in imaginal discs, where it acts as an Epidermal Growth Factor Receptor repressor. Our study suggests that the amount of Fasciclin 2 may determine a threshold to grow or stop growing during epithelial organ development.
Collapse
|
17
|
A role for Flower and cell death in controlling morphogen gradient scaling. Nat Cell Biol 2022; 24:424-433. [PMID: 35301437 DOI: 10.1038/s41556-022-00858-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 01/28/2022] [Indexed: 11/08/2022]
Abstract
During development, morphogen gradients encode positional information to pattern morphological structures during organogenesis1. Some gradients, like that of Dpp in the fly wing, remain proportional to the size of growing organs-that is, they scale. Gradient scaling keeps morphological patterns proportioned in organs of different sizes2,3. Here we show a mechanism of scaling that ensures that, when the gradient is smaller than the organ, cell death trims the developing tissue to match the size of the gradient. Scaling is controlled by molecular associations between Dally and Pentagone, known factors involved in scaling, and a key factor that mediates cell death, Flower4-6. We show that Flower activity in gradient expansion is not dominated by cell death, but by the activity of Dally/Pentagone on scaling. Here we show a potential connection between scaling and cell death that may uncover a molecular toolbox hijacked by tumours.
Collapse
|
18
|
Kietz C, Mohan AK, Pollari V, Tuominen IE, Ribeiro PS, Meier P, Meinander A. Drice restrains Diap2-mediated inflammatory signalling and intestinal inflammation. Cell Death Differ 2022; 29:28-39. [PMID: 34262145 PMCID: PMC8738736 DOI: 10.1038/s41418-021-00832-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The Drosophila IAP protein, Diap2, is a key mediator of NF-κB signalling and innate immune responses. Diap2 is required for both local immune activation, taking place in the epithelial cells of the gut and trachea, and for mounting systemic immune responses in the cells of the fat body. We have found that transgenic expression of Diap2 leads to a spontaneous induction of NF-κB target genes, inducing chronic inflammation in the Drosophila midgut, but not in the fat body. Drice is a Drosophila effector caspase known to interact and form a stable complex with Diap2. We have found that this complex formation induces its subsequent degradation, thereby regulating the amount of Diap2 driving NF-κB signalling in the intestine. Concordantly, loss of Drice activity leads to accumulation of Diap2 and to chronic intestinal inflammation. Interestingly, Drice does not interfere with pathogen-induced signalling, suggesting that it protects from immune responses induced by resident microbes. Accordingly, no inflammation was detected in transgenic Diap2 flies and Drice-mutant flies reared in axenic conditions. Hence, we show that Drice, by restraining Diap2, halts unwanted inflammatory signalling in the intestine.
Collapse
Affiliation(s)
- Christa Kietz
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Aravind K Mohan
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Vilma Pollari
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Ida-Emma Tuominen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland
| | - Paulo S Ribeiro
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, BioCity, Turku, Finland.
| |
Collapse
|
19
|
Killing by Degradation: Regulation of Apoptosis by the Ubiquitin-Proteasome-System. Cells 2021; 10:cells10123465. [PMID: 34943974 PMCID: PMC8700063 DOI: 10.3390/cells10123465] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022] Open
Abstract
Apoptosis is a cell suicide process that is essential for development, tissue homeostasis and human health. Impaired apoptosis is associated with a variety of human diseases, including neurodegenerative disorders, autoimmunity and cancer. As the levels of pro- and anti-apoptotic proteins can determine the life or death of cells, tight regulation of these proteins is critical. The ubiquitin proteasome system (UPS) is essential for maintaining protein turnover, which can either trigger or inhibit apoptosis. In this review, we will describe the E3 ligases that regulate the levels of pro- and anti-apoptotic proteins and assisting proteins that regulate the levels of these E3 ligases. We will provide examples of apoptotic cell death modulations using the UPS, determined by positive and negative feedback loop reactions. Specifically, we will review how the stability of p53, Bcl-2 family members and IAPs (Inhibitor of Apoptosis proteins) are regulated upon initiation of apoptosis. As increased levels of oncogenes and decreased levels of tumor suppressor proteins can promote tumorigenesis, targeting these pathways offers opportunities to develop novel anti-cancer therapies, which act by recruiting the UPS for the effective and selective killing of cancer cells.
Collapse
|
20
|
Beitel GJ. Organ remodeling: The molecular choreography of a dance of destruction. Curr Biol 2021; 31:R1517-R1520. [PMID: 34875239 DOI: 10.1016/j.cub.2021.10.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
A new study reveals that the Drosophila tracheal system is disassembled during pupation via ecdysone-dependent remodeling of the extracellular matrix, which then signals through the Hippo-Yorkie/YAP network to induce apoptosis.
Collapse
Affiliation(s)
- Greg J Beitel
- Beitel Lab, Northwestern University, Hogan Hall, Room 2-100, 2205 Tech Drive, Evanston, IL 60208-3500, USA.
| |
Collapse
|
21
|
Tilikj N, Novo M. How to resist soil desiccation: Transcriptional changes in a Mediterranean earthworm during aestivation. Comp Biochem Physiol A Mol Integr Physiol 2021; 264:111112. [PMID: 34748936 DOI: 10.1016/j.cbpa.2021.111112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/27/2022]
Abstract
Earthworms have a central role in ministering the terrestrial ecosystems and are proving to have an important role in modulating the effects climate change has on soil. Aestivation is a form of dormancy employed by the organisms living in deserts and arid environments, when confronted with prolonged periods of drought. Understanding global metabolic adjustments required for withstanding the harsh conditions of the ever more severe Iberian drought, we performed a global transcriptomic exploration of the endogeic earthworm Carpetania matritensis during aestivation. There were a total of 6352 differentially expressed transcripts in the aestivating group, with 65% being downregulated. Based on GO and KEGG enrichment analyses, downregulated genes seem to be indicative of an overall metabolic depression during aestivation. Indeed we noted a reduction of protein turnover and macromolecule metabolism coupled with suppression of genes involved in digestion. Upregulated genes, namely antioxidant genes and DNA repair genes showed clear signs of abiotic stress caused by ROS generation. Abiotic stress led to transcriptomic changes of genes involved in immune response, mostly affecting the NF-kb signaling pathway as well as changes in apoptotic genes indicating the necessity of investigating these processes in a tissue specific manner. Lastly we uncovered a possible mechanism for water retention by nitrogenous waste accumulation. This study provides the first ever transcriptomic investigation done on aestivating earthworms and as such serves as a general framework for investigation on other earthworm species and other soil invertebrates, which is becoming increasingly important with the current scenario of climate change.
Collapse
Affiliation(s)
- Natasha Tilikj
- Biodiversity, Ecology and Evolution Department, Faculty of Biology, Complutense University of Madrid, C/José Antonio Nováis 12, 28040 Madrid, Spain.
| | - Marta Novo
- Biodiversity, Ecology and Evolution Department, Faculty of Biology, Complutense University of Madrid, C/José Antonio Nováis 12, 28040 Madrid, Spain
| |
Collapse
|
22
|
Mehta AS, Deshpande P, Chimata AV, Tsonis PA, Singh A. Newt regeneration genes regulate Wingless signaling to restore patterning in Drosophila eye. iScience 2021; 24:103166. [PMID: 34746690 PMCID: PMC8551474 DOI: 10.1016/j.isci.2021.103166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/02/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Newts utilize their unique genes to restore missing parts by strategic regulation of conserved signaling pathways. Lack of genetic tools poses challenges to determine the function of such genes. Therefore, we used the Drosophila eye model to demonstrate the potential of 5 unique newt (Notophthalmus viridescens) gene(s), viropana1-viropana5 (vna1-vna5), which were ectopically expressed in L 2 mutant and GMR-hid, GMR-GAL4 eye. L 2 exhibits the loss of ventral half of early eye and head involution defective (hid) triggers cell-death during later eye development. Surprisingly, newt genes significantly restore missing photoreceptor cells both in L 2 and GMR>hid background by upregulating cell-proliferation and blocking cell-death, regulating evolutionarily conserved Wingless (Wg)/Wnt signaling pathway and exhibit non-cell-autonomous rescues. Further, Wg/Wnt signaling acts downstream of newt genes. Our data highlights that unique newt proteins can regulate conserved pathways to trigger a robust restoration of missing photoreceptor cells in Drosophila eye model with weak restoration capability.
Collapse
Affiliation(s)
| | | | | | | | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA
- Premedical Program, University of Dayton, Dayton, USA
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, USA
- The Integrative Science and Engineering Center, University of Dayton, Dayton, OH 45469, USA
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
23
|
Fraire-Zamora JJ, Tosi S, Solon J, Casanova J. Control of hormone-driven organ disassembly by ECM remodeling and Yorkie-dependent apoptosis. Curr Biol 2021; 31:5261-5273.e4. [PMID: 34666006 DOI: 10.1016/j.cub.2021.09.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 06/30/2021] [Accepted: 09/22/2021] [Indexed: 01/13/2023]
Abstract
Epithelia grow and shape into functional structures during organogenesis. Although most of the focus on organogenesis has been drawn to the building of biological structures, the disassembly of pre-existing structures is also an important event to reach a functional adult organ. Examples of disassembly processes include the regression of the Müllerian or Wolffian ducts during gonad development and mammary gland involution during the post-lactational period in adult females. To date, it is unclear how organ disassembly is controlled at the cellular level. Here, we follow the Drosophila larval trachea through metamorphosis and show that its disassembly is a hormone-driven and precisely orchestrated process. It occurs in two phases: first, remodeling of the apical extracellular matrix (aECM), mediated by matrix metalloproteases and independent of the actomyosin cytoskeleton, results in a progressive shortening of the entire trachea and a nuclear-to-cytoplasmic relocalization of the Hippo effector Yorkie (Yki). Second, a decreased transcription of the Yki target, Diap1, in the posterior metameres and the activation of caspases result in the apoptotic loss of the posterior half of the trachea while the anterior half escapes cell death. Thus, our work unravels a mechanism by which hormone-driven ECM remodeling controls sequential tissue shortening and apoptotic cell removal through the transcriptional activity of Yki, leading to organ disassembly during animal development.
Collapse
Affiliation(s)
- Juan J Fraire-Zamora
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Institut de Biologia Molecular de Barcelona (CSIC), Barcelona, Catalonia, Spain; Institut de Recerca Biomèdica (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain; Instituto Biofisika (CSIC, UPV/EHU), Basque Excellence Research Centre, Barrio Sarriena, 48940 Leioa, Spain.
| | - Sébastien Tosi
- Institut de Recerca Biomèdica (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Jérôme Solon
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain; Instituto Biofisika (CSIC, UPV/EHU), Basque Excellence Research Centre, Barrio Sarriena, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain.
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), Barcelona, Catalonia, Spain; Institut de Recerca Biomèdica (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| |
Collapse
|
24
|
Yang Y, Kong R, Goh FG, Somers WG, Hime GR, Li Z, Cai Y. dRTEL1 is essential for the maintenance of Drosophila male germline stem cells. PLoS Genet 2021; 17:e1009834. [PMID: 34644293 PMCID: PMC8513875 DOI: 10.1371/journal.pgen.1009834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/23/2021] [Indexed: 11/19/2022] Open
Abstract
Stem cells have the potential to maintain undifferentiated state and differentiate into specialized cell types. Despite numerous progress has been achieved in understanding stem cell self-renewal and differentiation, many fundamental questions remain unanswered. In this study, we identify dRTEL1, the Drosophila homolog of Regulator of Telomere Elongation Helicase 1, as a novel regulator of male germline stem cells (GSCs). Our genome-wide transcriptome analysis and ChIP-Seq results suggest that dRTEL1 affects a set of candidate genes required for GSC maintenance, likely independent of its role in DNA repair. Furthermore, dRTEL1 prevents DNA damage-induced checkpoint activation in GSCs. Finally, dRTEL1 functions to sustain Stat92E protein levels, the key player in GSC maintenance. Together, our findings reveal an intrinsic role of the DNA helicase dRTEL1 in maintaining male GSC and provide insight into the function of dRTEL1.
Collapse
Affiliation(s)
- Ying Yang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Ruiyan Kong
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Feng Guang Goh
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - W. Gregory Somers
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Gary R. Hime
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Hounsell C, Fan Y. The Duality of Caspases in Cancer, as Told through the Fly. Int J Mol Sci 2021; 22:8927. [PMID: 34445633 PMCID: PMC8396359 DOI: 10.3390/ijms22168927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Caspases, a family of cysteine-aspartic proteases, have an established role as critical components in the activation and initiation of apoptosis. Alongside this a variety of non-apoptotic caspase functions in proliferation, differentiation, cellular plasticity and cell migration have been reported. The activity level and context are important factors in determining caspase function. As a consequence of their critical role in apoptosis and beyond, caspases are uniquely situated to have pathological roles, including in cancer. Altered caspase function is a common trait in a variety of cancers, with apoptotic evasion defined as a "hallmark of cancer". However, the role that caspases play in cancer is much more complex, acting both to prevent and to promote tumourigenesis. This review focuses on the major findings in Drosophila on the dual role of caspases in tumourigenesis. This has major implications for cancer treatments, including chemotherapy and radiotherapy, with the activation of apoptosis being the end goal. However, such treatments may inadvertently have adverse effects on promoting tumour progression and acerbating the cancer. A comprehensive understanding of the dual role of caspases will aid in the development of successful cancer therapeutic approaches.
Collapse
Affiliation(s)
| | - Yun Fan
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
26
|
Bejarano F, Chang CH, Sun K, Hagen JW, Deng WM, Lai EC. A comprehensive in vivo screen for anti-apoptotic miRNAs indicates broad capacities for oncogenic synergy. Dev Biol 2021; 475:10-20. [PMID: 33662357 PMCID: PMC8107139 DOI: 10.1016/j.ydbio.2021.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
microRNAs (miRNAs) are ~21-22 nucleotide (nt) RNAs that mediate broad post-transcriptional regulatory networks. However, genetic analyses have shown that the phenotypic consequences of deleting individual miRNAs are generally far less overt compared to their misexpression. This suggests that miRNA deregulation may have broader phenotypic impacts during disease situations. We explored this concept in the Drosophila eye, by screening for miRNAs whose misexpression could modify the activity of pro-apoptotic factors. Via unbiased and comprehensive in vivo phenotypic assays, we identify an unexpectedly large set of miRNA hits that can suppress the action of pro-apoptotic genes hid and grim. We utilize secondary assays to validate that a subset of these miRNAs can inhibit irradiation-induced cell death. Since cancer cells might seek to evade apoptosis pathways, we modeled this situation by asking whether activation of anti-apoptotic miRNAs could serve as "second hits". Indeed, while clones of the lethal giant larvae (lgl) tumor suppressor are normally eliminated during larval development, we find that diverse anti-apoptotic miRNAs mediate the survival of lgl mutant clones in third instar larvae. Notably, while certain anti-apoptotic miRNAs can target apoptotic factors, most of our screen hits lack obvious targets in the core apoptosis machinery. These data highlight how a genetic approach can reveal distinct and powerful activities of miRNAs in vivo, including unexpected functional synergies during disease or cancer-relevant settings.
Collapse
Affiliation(s)
- Fernando Bejarano
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Ave, Box 252, New York, NY, 10065, USA
| | - Chih-Hsuan Chang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Kailiang Sun
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Ave, Box 252, New York, NY, 10065, USA; Weill Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Joshua W Hagen
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Ave, Box 252, New York, NY, 10065, USA; Tri-Institutional M.D.-Ph.D. Program, New York, NY, 10065, USA
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Ave, Box 252, New York, NY, 10065, USA.
| |
Collapse
|
27
|
Fan WM, Luo D, Zhang JZ, Wang D, Shen J. Vestigial suppresses apoptosis and cell migration in a manner dependent on the level of JNK-Caspase signaling in the Drosophila wing disc. INSECT SCIENCE 2021; 28:63-76. [PMID: 32037698 DOI: 10.1111/1744-7917.12762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/02/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
The Decapentaplegic (Dpp) and Wingless (Wg) signal pathways play important roles in numerous biological processes in Drosophila. The Drosophila vestigial (vg) gene is selectively required for wing imaginal disc cell proliferation, which is essential for the formation of the adult wing and halter structures, and is regulated by Dpp and Wg signaling. Using a Drosophila invasion model of wing epithelium, we showed herein that inhibition of Dpp or Wg signaling promoted cells to migrate across the cell lineage restrictive anterior/posterior (A/P) compartment boundary. Being downstream of both Dpp and Wg signaling, vg can block cell migration induced by loss of either pathway. In addition, suppression of vg is sufficient to induce cell migration across the A/P boundary. Transcriptomic analysis revealed potential downstream genes involved in the cell migration after suppressing vg in the wing disc. We further demonstrated that the c-Jun N-terminal kinase (JNK) signaling promoted cell migration induced by vg suppression by upregulating Caspase activity. Taken together, our results revealed the requirement of Vg for suppressing cell migration and clarified how developmental signals collaborate to stabilize cells along the compartment boundary.
Collapse
Affiliation(s)
- Wen-Min Fan
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Dan Luo
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Jun-Zheng Zhang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| |
Collapse
|
28
|
Lee G, Park JH. Programmed cell death reshapes the central nervous system during metamorphosis in insects. CURRENT OPINION IN INSECT SCIENCE 2021; 43:39-45. [PMID: 33065339 PMCID: PMC10754214 DOI: 10.1016/j.cois.2020.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/08/2020] [Accepted: 09/29/2020] [Indexed: 06/11/2023]
Abstract
Metamorphosis is fascinating and dramatic stage of postembryonic development in insects [1]. The most prominent metamorphic changes seen in holometabolous insects involve destruction of most larval structures and concomitant generation of adult ones. Such diverse cellular events are orchestrated by ecdysone. The central nervous system (CNS) is also extensively remodeled to process new sensory inputs; to coordinate new types of locomotion; and to perform higher-order decision making [2]. Programmed cell death (PCD) is an integral part of the metamorphic development. It eliminates obsolete larval tissues and extra cells that are generated from the morphogenesis of adult tissues. In the CNS, PCD of selected neurons and glial cells as well as reshaping of persistent larval cells are essential for establishing the adult CNS. In this review, we summarize the ecdysone signaling, and then molecular and cellular events associated with PCD primarily in the metamorphosing CNS of Drosophila melanogaster.
Collapse
Affiliation(s)
- Gyunghee Lee
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville TN 37996, United States
| | - Jae H Park
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville TN 37996, United States.
| |
Collapse
|
29
|
Savadogo EH, Shiomi Y, Yasuda J, Akino T, Yamaguchi M, Yoshida H, Umegawachi T, Tanaka R, Suong DNA, Miura K, Yazaki K, Kitajima S. Gene expression of PLAT and ATS3 proteins increases plant resistance to insects. PLANTA 2021; 253:37. [PMID: 33464406 DOI: 10.1007/s00425-020-03530-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Genes of the PLAT protein family, including PLAT and ATS3 subfamilies of higher plants and homologs of liverwort, are involved in plant defense against insects. Laticifer cells in plants contain large amounts of anti-microbe or anti-insect proteins and are involved in plant defense against biotic stresses. We previously found that PLAT proteins accumulate in laticifers of fig tree (Ficus carica) at comparable levels to those of chitinases, and the transcript level of ATS3, another PLAT domain-containing protein, is highest in the transcriptome of laticifers of Euphorbia tirucalli. In this study, we investigated whether the PLAT domain-containing proteins are involved in defense against insects. Larvae of the lepidopteran Spodoptera litura showed retarded growth when fed with Nicotiana benthamiana leaves expressing F. carica PLAT or E. tirucalli ATS3 genes, introduced by agroinfiltration using expression vector pBYR2HS. Transcriptome analysis of these leaves indicated that ethylene and jasmonate signaling were activated, leading to increased expression of genes for PR-1, β-1,3-glucanase, PR5 and trypsin inhibitors, suggesting an indirect mechanism of PLAT- and ATS3-induced resistance in the host plant. Direct cytotoxicity of PLAT and ATS3 to insects was also possible because heterologous expression of the corresponding genes in Drosophila melanogaster caused apoptosis-mediated cell death in this insect. Larval growth retardation of S. litura occurred when they were fed radish sprouts, a good host for agroinfiltration, expressing any of nine homologous genes of dicotyledon Arabidopsis thaliana, monocotyledon Brachypodium distachyon, conifer Picea sitchensis and liverwort Marchantia polymorpha. Of these nine genes, the heterologous expression of A. thaliana AT5G62200 and AT5G62210 caused significant increases in larval death. These results indicated that the PLAT protein family has largely conserved anti-insect activity in the plant kingdom (249 words).
Collapse
Affiliation(s)
- Eric Hyrmeya Savadogo
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yui Shiomi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Junko Yasuda
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Toshiharu Akino
- The Center for Advanced Insect Research Promotion, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Takanari Umegawachi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Ryo Tanaka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Dang Ngoc Anh Suong
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan
| | - Kenji Miura
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Japan
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Japan
| | - Kazufumi Yazaki
- Research Institute for Sustainable Humanosphere, Kyoto University, Uji, 611-0011, Japan
| | - Sakihito Kitajima
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan.
- The Center for Advanced Insect Research Promotion, Kyoto Institute of Technology, Matsugasaki Sakyo-ku, Kyoto, 606-8585, Japan.
| |
Collapse
|
30
|
DeAngelis MW, Coolon JD, Johnson RI. Comparative transcriptome analyses of the Drosophila pupal eye. G3-GENES GENOMES GENETICS 2021; 11:5995320. [PMID: 33561221 PMCID: PMC8043229 DOI: 10.1093/g3journal/jkaa003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/08/2020] [Indexed: 12/04/2022]
Abstract
Tissue function is dependent on correct cellular organization and behavior. As a result, the identification and study of genes that contribute to tissue morphogenesis is of paramount importance to the fields of cell and developmental biology. Many of the genes required for tissue patterning and organization are highly conserved between phyla. This has led to the emergence of several model organisms and developmental systems that are used to study tissue morphogenesis. One such model is the Drosophila melanogaster pupal eye that has a highly stereotyped arrangement of cells. In addition, the pupal eye is postmitotic that allows for the study of tissue morphogenesis independent from any effects of proliferation. While the changes in cell morphology and organization that occur throughout pupal eye development are well documented, less is known about the corresponding transcriptional changes that choreograph these processes. To identify these transcriptional changes, we dissected wild-type Canton S pupal eyes and performed RNA-sequencing. Our analyses identified differential expression of many loci that are documented regulators of pupal eye morphogenesis and contribute to multiple biological processes including signaling, axon projection, adhesion, and cell survival. We also identified differential expression of genes not previously implicated in pupal eye morphogenesis such as components of the Toll pathway, several non-classical cadherins, and components of the muscle sarcomere, which could suggest these loci function as novel patterning factors.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Joseph D Coolon
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Ruth I Johnson
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| |
Collapse
|
31
|
Abstract
Apoptotic processes play an important role in the development and physiology of almost all metazoan clades. In the highly diverse group of insects, apoptotic pathways have been characterized in only a few dipteran and lepidopteran species, which may not be representative of all insect species. Here, we report the first complete annotation of the apoptotic pathway in a hemipteran insect, the pea aphid Acyrthosiphon pisum. We showed that its apoptotic pathway is rewired compared to other insects, with a significant increase in the number of inhibitors of apoptosis (IAPs) and evidence for functional diversification and structural modularity of this protein family. These novelties are widespread in the aphid lineage, suggesting a yet not understood novel aphid-specific function of IAPs. Apoptosis, a conserved form of programmed cell death, shows interspecies differences that may reflect evolutionary diversification and adaptation, a notion that remains largely untested. Among insects, the most speciose animal group, the apoptotic pathway has only been fully characterized in Drosophila melanogaster, and apoptosis-related proteins have been studied in a few other dipteran and lepidopteran species. Here, we studied the apoptotic pathway in the aphid Acyrthosiphon pisum, an insect pest belonging to the Hemiptera, an earlier-diverging and distantly related order. We combined phylogenetic analyses and conserved domain identification to annotate the apoptotic pathway in A. pisum and found low caspase diversity and a large expansion of its inhibitory part, with 28 inhibitors of apoptosis (IAPs). We analyzed the spatiotemporal expression of a selected set of pea aphid IAPs and showed that they are differentially expressed in different life stages and tissues, suggesting functional diversification. Five IAPs are specifically induced in bacteriocytes, the specialized cells housing symbiotic bacteria, during their cell death. We demonstrated the antiapoptotic role of these five IAPs using heterologous expression in a tractable in vivo model, the Drosophila melanogaster developing eye. Interestingly, IAPs with the strongest antiapoptotic potential contain two BIR and two RING domains, a domain association that has not been observed in any other species. We finally analyzed all available aphid genomes and found that they all show large IAP expansion, with new combinations of protein domains, suggestive of evolutionarily novel aphid-specific functions.
Collapse
|
32
|
Ai X, Wang D, Zhang J, Shen J. Hippo signaling promotes Ets21c-dependent apical cell extrusion in the Drosophila wing disc. Development 2020; 147:dev.190124. [PMID: 33028612 DOI: 10.1242/dev.190124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/28/2020] [Indexed: 01/11/2023]
Abstract
Cell extrusion is a crucial regulator of epithelial tissue development and homeostasis. Epithelial cells undergoing apoptosis, bearing pathological mutations or possessing developmental defects are actively extruded toward elimination. However, the molecular mechanisms of Drosophila epithelial cell extrusion are not fully understood. Here, we report that activation of the conserved Hippo (Hpo) signaling pathway induces both apical and basal cell extrusion in the Drosophila wing disc epithelia. We show that canonical Yorkie targets Diap1, Myc and Cyclin E are not required for either apical or basal cell extrusion induced by activation of this pathway. Another target gene, bantam, is only involved in basal cell extrusion, suggesting novel Hpo-regulated apical cell extrusion mechanisms. Using RNA-seq analysis, we found that JNK signaling is activated in the extruding cells. We provide genetic evidence that JNK signaling activation is both sufficient and necessary for Hpo-regulated cell extrusion. Furthermore, we demonstrate that the ETS-domain transcription factor Ets21c, an ortholog of proto-oncogenes FLI1 and ERG, acts downstream of JNK signaling to mediate apical cell extrusion. Our findings reveal a novel molecular link between Hpo signaling and cell extrusion.
Collapse
Affiliation(s)
- Xianlong Ai
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing 100193, China
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing 100193, China
| | - Junzheng Zhang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing 100193, China
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, China Agricultural University, Beijing 100193, China
| |
Collapse
|
33
|
Chereddy SCRR, Gurusamy D, Howell JL, Palli SR. Double-stranded RNAs targeting inhibitor of apoptosis gene show no significant cross-species activity. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 104:e21683. [PMID: 32350930 PMCID: PMC9987616 DOI: 10.1002/arch.21683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/19/2020] [Accepted: 04/02/2020] [Indexed: 05/06/2023]
Abstract
RNA interference (RNAi) has become an integral part of mainstream research due to its versatility and ease of use. However, the potential nontarget effects associated with double-stranded RNAs (dsRNA) are poorly understood. To explore this, we used dsRNAs targeting the inhibitor of apoptosis (iap) gene from nine insect species and assayed their possible nontarget effects. For each assay, we used a control (dsRNA targeting the gene coding for green fluorescent protein, GFP) and a species-specific dsRNA targeting nine iap genes in insect species to evaluate target gene knockdown efficiency, apoptosis phenotype in cells and mortality in insects. Our results revealed that dsIAP efficiently knocks down iap gene expression and induces apoptosis phenotype and mortality in target insect species. In contrast, no significant knockdown of the iap gene expression, apoptosis phenotypes, or mortality were detected in cell lines developed from nontarget insects or nontarget insects treated with dsIAPs. Interestingly, even among closely related insects such as stink bugs, Nezara viridula, Halyomorpha halys, and Murgantia histrionica, with substantial sequence similarity among iap genes from these insects, no significant nontarget effects of dsIAP were observed under the conditions tested. These data demonstrate no significant nontarget effects for dsIAPs and suggest that the threat of nontarget effects of RNAi technology may not be substantial.
Collapse
Affiliation(s)
| | | | - Jeffrey L Howell
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| | - Subba R Palli
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
34
|
Yoon JS, Koo J, George S, Palli SR. Evaluation of inhibitor of apoptosis genes as targets for RNAi-mediated control of insect pests. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 104:e21689. [PMID: 32394607 PMCID: PMC9945918 DOI: 10.1002/arch.21689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/10/2020] [Accepted: 04/21/2020] [Indexed: 05/12/2023]
Abstract
Apoptosis has been widely studied from mammals to insects. Inhibitor of apoptosis (IAP) protein is a negative regulator of apoptosis. Recent studies suggest that iap genes could be excellent targets for RNA interference (RNAi)-mediated control of insect pests. However, not much is known about iap genes in one of the well-known insect model species, Tribolium castaneum. The orthologues of five iap genes were identified in T. castaneum by searching its genome at NCBI (https://www.ncbi.nlm.nih.gov/) and UniProt (https://www.uniprot.org/) databases using Drosophila melanogaster and Aedes aegypti IAP protein sequences as queries. RNAi assays were performed in T. castaneum cell line (TcA) and larvae. The knockdown of iap1 gene induced a distinct apoptotic phenotype in TcA cells and induced 91% mortality in T. castaneum larvae. Whereas, knockdown of iap5 resulted in a decrease in cell proliferation in TcA cells and developmental defects in T. castaneum larvae which led to 100% mortality. Knockdown of the other three iap genes identified did not cause a significant effect on cells or insects. These data increase our understanding of iap genes in insects and provide opportunities for developing iap1 and iap5 as targets for RNAi-based insect pest control.
Collapse
Affiliation(s)
- June-Sun Yoon
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| | - Jinmo Koo
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| | - Smitha George
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| | - Subba R Palli
- Department of Entomology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
35
|
DeAngelis MW, McGhie EW, Coolon JD, Johnson RI. Mask, a component of the Hippo pathway, is required for Drosophila eye morphogenesis. Dev Biol 2020; 464:53-70. [PMID: 32464117 DOI: 10.1016/j.ydbio.2020.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022]
Abstract
Hippo signaling is an important regulator of tissue size, but it also has a lesser-known role in tissue morphogenesis. Here we use the Drosophila pupal eye to explore the role of the Hippo effector Yki and its cofactor Mask in morphogenesis. We found that Mask is required for the correct distribution and accumulation of adherens junctions and appropriate organization of the cytoskeleton. Accordingly, disrupting mask expression led to severe mis-patterning and similar defects were observed when yki was reduced or in response to ectopic wts. Further, the patterning defects generated by reducing mask expression were modified by Hippo pathway activity. RNA-sequencing revealed a requirement for Mask for appropriate expression of numerous genes during eye morphogenesis. These included genes implicated in cell adhesion and cytoskeletal organization, a comprehensive set of genes that promote cell survival, and numerous signal transduction genes. To validate our transcriptome analyses, we then considered two loci that were modified by Mask activity: FER and Vinc, which have established roles in regulating adhesion. Modulating the expression of either locus modified mask mis-patterning and adhesion phenotypes. Further, expression of FER and Vinc was modified by Yki. It is well-established that the Hippo pathway is responsive to changes in cell adhesion and the cytoskeleton, but our data indicate that Hippo signaling also regulates these structures.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Emily W McGhie
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Joseph D Coolon
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Ruth I Johnson
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| |
Collapse
|
36
|
Ciao1 interacts with Crumbs and Xpd to regulate organ growth in Drosophila. Cell Death Dis 2020; 11:365. [PMID: 32404863 PMCID: PMC7220951 DOI: 10.1038/s41419-020-2564-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 11/09/2022]
Abstract
Ciao1 is a component of the cytosolic iron-sulfur cluster assembly (CIA) complex along with MMS19 and MIP18. Xeroderma pigmentosum group D (XPD), a DNA helicase involved in regulation of cell cycle and transcription, is a CIA target for iron-sulfur (Fe/S) modification. In vivo function of Ciao1 and Xpd in developing animals has been rarely studied. Here, we reveal that Ciao1 interacts with Crumbs (Crb), Galla, and Xpd to regulate organ growth in Drosophila. Abnormal growth of eye by overexpressing Crb intracellular domain (Crbintra) is suppressed by reducing the Ciao1 level. Loss of Ciao1 or Xpd causes similar impairment in organ growth. RNAi knockdown of both Ciao1 and Xpd show similar phenotypes as Ciao1 or Xpd RNAi alone, suggesting their function in a pathway. Growth defects caused by Ciao1 RNAi are suppressed by overexpression of Xpd. Ciao1 physically interacts with Crbintra, Galla, and Xpd, supporting their genetic interactions. Remarkably, Xpd RNAi defects can also be suppressed by Ciao1 overexpression, implying a mutual regulation between the two genes. Ciao1 mutant clones in imaginal discs show decreased levels of Cyclin E (CycE) and death-associated inhibitor of apoptosis 1 (Diap1). Xpd mutant clones share the similar reduction of CycE and Diap1. Consequently, knockdown of Ciao1 and Xpd by RNAi show increased apoptotic cell death. Further, CycE overexpression is sufficient to restore the growth defects from Ciao1 RNAi or Xpd RNAi. Interestingly, Diap1 overexpression in Ciao1 mutant clones induces CycE expression, suggesting that reduced CycE in Ciao1 mutant cells is secondary to loss of Diap1. Taken together, this study reveals new roles of Ciao1 and Xpd in cell survival and growth through regulating Diap1 level during organ development.
Collapse
|
37
|
Myc plays an important role in Drosophila P-M hybrid dysgenesis to eliminate germline cells with genetic damage. Commun Biol 2020; 3:185. [PMID: 32322015 PMCID: PMC7176646 DOI: 10.1038/s42003-020-0923-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/24/2020] [Indexed: 11/08/2022] Open
Abstract
Genetic damage in the germline induced by P-element mobilization causes a syndrome known as P-M hybrid dysgenesis (HD), which manifests as elevated mutation frequency and loss of germline cells. In this study, we found that Myc plays an important role in eliminating germline cells in the context of HD. P-element mobilization resulted in downregulation of Myc expression in the germline. Myc knockdown caused germline elimination; conversely, Myc overexpression rescued the germline loss caused by P-element mobilization. Moreover, restoration of fertility by Myc resulted in the production of gametes with elevated mutation frequency and reduced ability to undergo development. Our results demonstrate that Myc downregulation mediates elimination of germline cells with accumulated genetic damage, and that failure to remove these cells results in increased production of aberrant gametes. Therefore, we propose that elimination of germline cells mediated by Myc downregulation is a quality control mechanism that maintains the genomic integrity of the germline.
Collapse
|
38
|
Hoshika S, Sun X, Kuranaga E, Umetsu D. Reduction of endocytic activity accelerates cell elimination during tissue remodeling of the Drosophila epidermal epithelium. Development 2020; 147:dev.179648. [PMID: 32156754 DOI: 10.1242/dev.179648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022]
Abstract
Epithelial tissues undergo cell turnover both during development and for homeostatic maintenance. Cells that are no longer needed are quickly removed without compromising the barrier function of the tissue. During metamorphosis, insects undergo developmentally programmed tissue remodeling. However, the mechanisms that regulate this rapid tissue remodeling are not precisely understood. Here, we show that the temporal dynamics of endocytosis modulate physiological cell properties to prime larval epidermal cells for cell elimination. Endocytic activity gradually reduces as tissue remodeling progresses. This reduced endocytic activity accelerates cell elimination through the regulation of Myosin II subcellular reorganization, junctional E-cadherin levels, and caspase activation. Whereas the increased Myosin II dynamics accelerates cell elimination, E-cadherin plays a protective role against cell elimination. Reduced E-cadherin is involved in the amplification of caspase activation by forming a positive-feedback loop with caspase. These findings reveal the role of endocytosis in preventing cell elimination and in the cell-property switching initiated by the temporal dynamics of endocytic activity to achieve rapid cell elimination during tissue remodeling.
Collapse
Affiliation(s)
- Shinichiro Hoshika
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Xiaofei Sun
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
39
|
Davidson AJ, Wood W. Phagocyte Responses to Cell Death in Flies. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036350. [PMID: 31501193 DOI: 10.1101/cshperspect.a036350] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multicellular organisms are not created through cell proliferation alone. It is through cell death that an indefinite cellular mass is pared back to reveal its true form. Cells are also lost throughout life as part of homeostasis and through injury. This detritus represents a significant burden to the living organism and must be cleared, most notably through the use of specialized phagocytic cells. Our understanding of these phagocytes and how they engulf cell corpses has been greatly aided by studying the fruit fly, Drosophila melanogaster Here we review the contribution of Drosophila research to our understanding of how phagocytes respond to cell death. We focus on the best studied phagocytes in the fly: the glia of the central nervous system, the ovarian follicle cells, and the macrophage-like hemocytes. Each is explored in the context of the tissue they maintain as well as how they function during development and in response to injury.
Collapse
Affiliation(s)
- Andrew J Davidson
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| | - Will Wood
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
40
|
Abbas R, Larisch S. Targeting XIAP for Promoting Cancer Cell Death-The Story of ARTS and SMAC. Cells 2020; 9:E663. [PMID: 32182843 PMCID: PMC7140716 DOI: 10.3390/cells9030663] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Inhibitors of apoptosis (IAPs) are a family of proteins that regulate cell death and inflammation. XIAP (X-linked IAP) is the only family member that suppresses apoptosis by directly binding to and inhibiting caspases. On the other hand, cIAPs suppress the activation of the extrinsic apoptotic pathway by preventing the formation of pro-apoptotic signaling complexes. IAPs are negatively regulated by IAP-antagonist proteins such as Smac/Diablo and ARTS. ARTS can promote apoptosis by binding and degrading XIAP via the ubiquitin proteasome-system (UPS). Smac can induce the degradation of cIAPs but not XIAP. Many types of cancer overexpress IAPs, thus enabling tumor cells to evade apoptosis. Therefore, IAPs, and in particular XIAP, have become attractive targets for cancer therapy. In this review, we describe the differences in the mechanisms of action between Smac and ARTS, and we summarize efforts to develop cancer therapies based on mimicking Smac and ARTS. Several Smac-mimetic small molecules are currently under evaluation in clinical trials. Initial efforts to develop ARTS-mimetics resulted in a novel class of compounds, which bind and degrade XIAP but not cIAPs. Smac-mimetics can target tumors with high levels of cIAPs, whereas ARTS-mimetics are expected to be effective for cancers with high levels of XIAP.
Collapse
Affiliation(s)
| | - Sarit Larisch
- Laboratory of Cell Death and Cancer Research, Biology& Human Biology Departments, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel;
| |
Collapse
|
41
|
Decoupling of Apoptosis from Activation of the ER Stress Response by the Drosophila Metallopeptidase superdeath. Genetics 2020; 214:913-925. [PMID: 32047096 DOI: 10.1534/genetics.119.303004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/09/2020] [Indexed: 12/19/2022] Open
Abstract
Endoplasmic reticulum (ER) stress-induced apoptosis is a primary cause and modifier of degeneration in a number of genetic disorders. Understanding how genetic variation influences the ER stress response and subsequent activation of apoptosis could improve individualized therapies and predictions of outcomes for patients. In this study, we find that the uncharacterized, membrane-bound metallopeptidase CG14516 in Drosophila melanogaster, which we rename as SUPpressor of ER stress-induced DEATH (su per death), plays a role in modifying ER stress-induced apoptosis. We demonstrate that loss of su per death reduces apoptosis and degeneration in the Rh1G69D model of ER stress through the JNK signaling cascade. This effect on apoptosis occurs without altering the activation of the unfolded protein response (IRE1 and PERK), suggesting that the beneficial prosurvival effects of this response are intact. Furthermore, we show that su per death functions epistatically upstream of CDK5-a known JNK-activated proapoptotic factor in this model of ER stress. We demonstrate that su per death is not only a modifier of this particular model, but affects the general tolerance to ER stress, including ER stress-induced apoptosis. Finally, we present evidence of Superdeath localization to the ER membrane. While similar in sequence to a number of human metallopeptidases found in the plasma membrane and ER membrane, its localization suggests that su per death is orthologous to ERAP1/2 in humans. Together, this study provides evidence that su per death is a link between stress in the ER and activation of cytosolic apoptotic pathways.
Collapse
|
42
|
Klipa O, Hamaratoglu F. Cell elimination strategies upon identity switch via modulation of apterous in Drosophila wing disc. PLoS Genet 2019; 15:e1008573. [PMID: 31877129 PMCID: PMC6952109 DOI: 10.1371/journal.pgen.1008573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 12/17/2019] [Indexed: 12/04/2022] Open
Abstract
The ability to establish spatial organization is an essential feature of any developing tissue and is achieved through well-defined rules of cell-cell communication. Maintenance of this organization requires elimination of cells with inappropriate positional identity, a poorly understood phenomenon. Here we studied mechanisms regulating cell elimination in the context of a growing tissue, the Drosophila wing disc and its dorsal determinant Apterous. Systematic analysis of apterous mutant clones along with their twin spots shows that they are eliminated from the dorsal compartment via three different mechanisms: relocation to the ventral compartment, basal extrusion, and death, depending on the position of the clone in the wing disc. We find that basal extrusion is the main elimination mechanism in the hinge, whereas apoptosis dominates in the pouch and in the notum. In the absence of apoptosis, extrusion takes over to ensure clearance in all regions. Notably, clones in the hinge grow larger than those in the pouch, emphasizing spatial differences. Mechanistically, we find that limiting cell division within the clones does not prevent their extrusion. Indeed, even clones of one or two cells can be extruded basally, demonstrating that the clone size is not the main determinant of the elimination mechanism to be used. Overall, we revealed three elimination mechanisms and their spatial biases for preserving pattern in a growing organ. As development proceeds, cells become more specialized and the compartmentalization ensures spatial separation of the specialized cells. This process of pattern formation is rather well understood. How the pattern is maintained afterwards though is largely unknown. Using the Drosophila wing disc as a model organ, we examined what happens to dorsal cells if they lose their dorsal identity. Formerly, it was shown that these cells are eliminated from the dorsal compartment via apoptosis or through relocation to the ventral compartment. Here we show that a third mode of elimination, basal extrusion, also contributes to their clearing. We quantified, for the first time, contributions of each mechanism and discovered a regional bias in their operation. Importantly, if apoptosis is blocked, basal extrusion takes over to ensure clearance from all regions. Recent modeling approaches suggested that there is a lower limit to the clone size for extrusion. Therefore, we tested the hypothesis that the choice of elimination mechanism may be dictated by the clone size. We prevented cell divisions within the clones to be eliminated and found that even 1–2 cell clones readily underwent basal extrusion, demonstrating that there is no lower limit to the clone size for extrusion.
Collapse
Affiliation(s)
- Olga Klipa
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Fisun Hamaratoglu
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Coelho DS, Schwartz S, Merino MM, Hauert B, Topfel B, Tieche C, Rhiner C, Moreno E. Culling Less Fit Neurons Protects against Amyloid-β-Induced Brain Damage and Cognitive and Motor Decline. Cell Rep 2019; 25:3661-3673.e3. [PMID: 30590040 PMCID: PMC6315112 DOI: 10.1016/j.celrep.2018.11.098] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/05/2018] [Accepted: 11/28/2018] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, impairing cognitive and motor functions. One of the pathological hallmarks of AD is neuronal loss, which is not reflected in mouse models of AD. Therefore, the role of neuronal death is still uncertain. Here, we used a Drosophila AD model expressing a secreted form of human amyloid-β42 peptide and showed that it recapitulates key aspects of AD pathology, including neuronal death and impaired long-term memory. We found that neuronal apoptosis is mediated by cell fitness-driven neuronal culling, which selectively eliminates impaired neurons from brain circuits. We demonstrated that removal of less fit neurons delays β-amyloid-induced brain damage and protects against cognitive and motor decline, suggesting that contrary to common knowledge, neuronal death may have a beneficial effect in AD. Peptides linked to neurodegenerative diseases reduce neuronal fitness in Drosophila β-amyloid-induced neuronal death is mediated by fitness regulators flower and azot Suppression of fitness-based neuronal culling aggravates cognitive and motor decline Neuronal death related to fitness-based selection has a beneficial net effect
Collapse
Affiliation(s)
- Dina S Coelho
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Silvia Schwartz
- Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Marisa M Merino
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Department of Biochemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Barbara Hauert
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Barbara Topfel
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Colin Tieche
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Christa Rhiner
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Eduardo Moreno
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|
44
|
Palu RAS, Ong E, Stevens K, Chung S, Owings KG, Goodman AG, Chow CY. Natural Genetic Variation Screen in Drosophila Identifies Wnt Signaling, Mitochondrial Metabolism, and Redox Homeostasis Genes as Modifiers of Apoptosis. G3 (BETHESDA, MD.) 2019; 9:3995-4005. [PMID: 31570502 PMCID: PMC6893197 DOI: 10.1534/g3.119.400722] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022]
Abstract
Apoptosis is the primary cause of degeneration in a number of neuronal, muscular, and metabolic disorders. These diseases are subject to a great deal of phenotypic heterogeneity in patient populations, primarily due to differences in genetic variation between individuals. This creates a barrier to effective diagnosis and treatment. Understanding how genetic variation influences apoptosis could lead to the development of new therapeutics and better personalized treatment approaches. In this study, we examine the impact of the natural genetic variation in the Drosophila Genetic Reference Panel (DGRP) on two models of apoptosis-induced retinal degeneration: overexpression of p53 or reaper (rpr). We identify a number of known apoptotic, neural, and developmental genes as candidate modifiers of degeneration. We also use Gene Set Enrichment Analysis (GSEA) to identify pathways that harbor genetic variation that impact these apoptosis models, including Wnt signaling, mitochondrial metabolism, and redox homeostasis. Finally, we demonstrate that many of these candidates have a functional effect on apoptosis and degeneration. These studies provide a number of avenues for modifying genes and pathways of apoptosis-related disease.
Collapse
Affiliation(s)
- Rebecca A S Palu
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Elaine Ong
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Kaitlyn Stevens
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Shani Chung
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Katie G Owings
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Alan G Goodman
- School of Molecular Biosciences, and
- Paul G. Allen School for Global Animal Health, Washington State University College of Veterinary Medicine, Pullman, WA 99164
| | - Clement Y Chow
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112,
| |
Collapse
|
45
|
Millet-Boureima C, Chingle R, Lubell WD, Gamberi C. Cyst Reduction in a Polycystic Kidney Disease Drosophila Model Using Smac Mimics. Biomedicines 2019; 7:biomedicines7040082. [PMID: 31635379 PMCID: PMC6966561 DOI: 10.3390/biomedicines7040082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited malady affecting 12.5 million people worldwide. Therapeutic options to treat PKD are limited, due in part to lack of precise knowledge of underlying pathological mechanisms. Mimics of the second mitochondria-derived activator of caspases (Smac) have exhibited activity as antineoplastic agents and reported recently to ameliorate cysts in a murine ADPKD model, possibly by differentially targeting cystic cells and sparing the surrounding tissue. A first-in-kind Drosophila PKD model has now been employed to probe further the activity of novel Smac mimics. Substantial reduction of cystic defects was observed in the Malpighian (renal) tubules of treated flies, underscoring mechanistic conservation of the cystic pathways and potential for efficient testing of drug prototypes in this PKD model. Moreover, the observed differential rescue of the anterior and posterior tubules overall, and within their physiologically diverse intermediate and terminal regions implied a nuanced response in distinct tubular regions contingent upon the structure of the Smac mimic. Knowledge gained from studying Smac mimics reveals the capacity for the Drosophila model to precisely probe PKD pharmacology highlighting the value for such critical evaluation of factors implicated in renal function and pathology.
Collapse
Affiliation(s)
| | - Ramesh Chingle
- Département de Chimie, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - William D Lubell
- Département de Chimie, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
46
|
Wang LH, Baker NE. Salvador-Warts-Hippo pathway regulates sensory organ development via caspase-dependent nonapoptotic signaling. Cell Death Dis 2019; 10:669. [PMID: 31511495 PMCID: PMC6739336 DOI: 10.1038/s41419-019-1924-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/03/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022]
Abstract
The fundamental roles for the Salvador-Warts-Hippo (SWH) pathway are widely characterized in growth regulation and organ size control. However, the function of SWH pathway is less known in cell fate determination. Here we uncover a novel role of the SWH signaling pathway in determination of cell fate during neural precursor (sensory organ precursor, SOP) development. Inactivation of the SWH pathway in SOP of the wing imaginal discs affects caspase-dependent bristle patterning in an apoptosis-independent process. Such nonapoptotic functions of caspases have been implicated in inflammation, proliferation, cellular remodeling, and cell fate determination. Our data indicate an effect on the Wingless (Wg)/Wnt pathway. Previously, caspases were proposed to cleave and activate a negative regulator of Wg/Wnt signaling, Shaggy (Sgg)/GSK3β. Surprisingly, we found that a noncleavable form of Sgg encoded from the endogenous locus after CRISPR-Cas9 modification supported almost normal bristle patterning, indicating that Sgg might not be the main target of the caspase-dependent nonapoptotic process. Collectively, our results outline a new function of SWH signaling that crosstalks to caspase-dependent nonapoptotic signaling and Wg/Wnt signaling in neural precursor development, which might be implicated in neuronal pathogenesis.
Collapse
Affiliation(s)
- Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161 Sec 6, Minquan E. Rd, Taipei, 11490, Taiwan.
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
47
|
Li M, Sun S, Priest J, Bi X, Fan Y. Characterization of TNF-induced cell death in Drosophila reveals caspase- and JNK-dependent necrosis and its role in tumor suppression. Cell Death Dis 2019; 10:613. [PMID: 31409797 PMCID: PMC6692325 DOI: 10.1038/s41419-019-1862-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/26/2022]
Abstract
Tumor-necrosis factor (TNF) and its superfamily members are pleiotropic cytokines. Activation of TNF can lead to distinct cellular outcomes including inflammation, cell survival, and different forms of cell death, such as apoptosis and necrosis in a context-dependent manner. However, our understanding of what determines the versatile functions of TNF is far from complete. Here, we examined the molecular mechanisms that distinguish the forms of cell death induced by Eiger (Egr), the sole homolog of TNF in Drosophila. We show that expression of Egr in the developing Drosophila eye simultaneously induces apoptosis and apoptosis-independent developmental defects indicated by cellular disorganization, both of which rely on the c-Jun N-terminal kinase (JNK) signaling activity. Intriguingly, when effector caspases DrICE and Dcp-1 are defective or inhibited, expression of Egr triggers necrosis which is characterized by loss of cell membrane integrity, translucent cytoplasm, and aggregation of cellular organelles. Moreover, such Egr-induced necrosis depends on the catalytic activity of the initiator caspase Dronc and the input from JNK signaling but is independent of their roles in apoptosis. Further mosaic analysis with mutants of scribble (scrib), an evolutionarily conserved tumor suppressor gene regulating cell polarity, suggests that Egr/JNK-mediated apoptosis and necrosis establish a two-layered defense system to inhibit the oncogenic growth of scrib mutant cells. Together, we have identified caspase- and JNK-dependent mechanisms underlying Egr-induced apoptosis versus necrosis and their fail-safe roles in tumor suppression in an intact organism in vivo.
Collapse
Affiliation(s)
- Mingli Li
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Shiyao Sun
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Jessica Priest
- School of Biosciences, University of Birmingham, Birmingham, UK
| | - Xiaolin Bi
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yun Fan
- School of Biosciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
48
|
The microRNA-306/abrupt regulatory axis controls wing and haltere growth in Drosophila. Mech Dev 2019; 158:103555. [PMID: 31112748 DOI: 10.1016/j.mod.2019.103555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Abstract
Growth control relies on extrinsic and intrinsic mechanisms that regulate and coordinate the size and pattern of organisms. This control is crucial for a homeostatic development and healthy physiology. The gene networks acting in this process are large and complex: factors involved in growth control are also important in diverse biological processes and these networks include multiple regulators that interact and respond to intra- and extra-cellular inputs that may ultimately converge in the control of the cell cycle. In this work we have studied the function of the Drosophila abrupt gene, coding for a BTB-ZF protein and previously reported to be required for wing vein pattern, in the control of haltere and wing growth. We have found that inactivation of abrupt reduces the size of the wing and haltere. We also found that the microRNA miR-306 controls abrupt expression and that miR-306 and abrupt genetically interact to control wing size. Moreover, the reduced appendage size due to abrupt inactivation is rescued by overexpression of Cyclin-E and by inactivation of dacapo. These findings define a miR-306-abrupt regulatory axis that controls wing and haltere size, whereby miR-306 maintains appropriate levels of abrupt expression which, in turn, regulates the cell cycle. Thus, our results uncover a novel function of abrupt in the regulation of the size of Drosophila appendages during development and contribute to the understanding of the coordination between growth and pattern as well as to the understanding of abrupt oncogenic function in flies.
Collapse
|
49
|
DeAngelis MW, Johnson RI. Dissection of the Drosophila Pupal Retina for Immunohistochemistry, Western Analysis, and RNA Isolation. J Vis Exp 2019. [PMID: 30933080 DOI: 10.3791/59299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The Drosophila pupal retina provides an excellent model system for the study of morphogenetic processes during development. In this paper, we present a reliable protocol for the dissection of the delicate Drosophila pupal retina. Our surgical approach utilizes readily-available microdissection tools to open pupae and precisely extract eye-brain complexes. These can be fixed, subjected to immunohistochemistry, and retinas then mounted onto microscope slides and imaged if the goal is to detect cellular or subcellular structures. Alternatively, unfixed retinas can be isolated from brain tissue, lysed in appropriate buffers and utilized for protein gel electrophoresis or mRNA extraction (to assess protein or gene expression, respectively). Significant practice and patience may be required to master the microdissection protocol described, but once mastered, the protocol enables relatively quick isolation of mainly undamaged retinas.
Collapse
|
50
|
The Caspase-3 homolog DrICE regulates endocytic trafficking during Drosophila tracheal morphogenesis. Nat Commun 2019; 10:1031. [PMID: 30833576 PMCID: PMC6399233 DOI: 10.1038/s41467-019-09009-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 02/13/2019] [Indexed: 12/30/2022] Open
Abstract
Although well known for its role in apoptosis, the executioner caspase DrICE has a non-apoptotic function that is required for elongation of the epithelial tubes of the Drosophila tracheal system. Here, we show that DrICE acts downstream of the Hippo Network to regulate endocytic trafficking of at least four cell polarity, cell junction and apical extracellular matrix proteins involved in tracheal tube size control: Crumbs, Uninflatable, Kune-Kune and Serpentine. We further show that tracheal cells are competent to undergo apoptosis, even though developmentally-regulated DrICE function rarely kills tracheal cells. Our results reveal a developmental role for caspases, a pool of DrICE that co-localizes with Clathrin, and a mechanism by which the Hippo Network controls endocytic trafficking. Given reports of in vitro regulation of endocytosis by mammalian caspases during apoptosis, we propose that caspase-mediated regulation of endocytic trafficking is an evolutionarily conserved function of caspases that can be deployed during morphogenesis. Caspases are well-known drivers of apoptosis, although recent studies suggest potential non-apoptotic functions. Here, McSharry and Beitel show that the Drosophila executioner caspase DrICE regulates endocytic trafficking of key proteins downstream of Hippo during tracheal morphogenesis.
Collapse
|