101
|
Lee SA, Jang SH, Kim BH, Shibata T, Yoo J, Jung Y, Kawabata SI, Lee BL. Insecticidal activity of the metalloprotease AprA occurs through suppression of host cellular and humoral immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 81:116-126. [PMID: 29174605 DOI: 10.1016/j.dci.2017.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/22/2017] [Accepted: 11/22/2017] [Indexed: 06/07/2023]
Abstract
The biochemical characterization of virulence factors from entomopathogenic bacteria is important to understand entomopathogen-insect molecular interactions. Pseudomonas entomophila is a typical entomopathogenic bacterium that harbors virulence factors against several insects. However, the molecular actions of these factors against host innate immune responses are not clearly elucidated. In this study, we observed that bean bugs (Riptortus pedestris) that were injected with P. entomophila were highly susceptible to this bacterium. To determine how P. entomophila counteracts the host innate immunity to survive within the insect, we purified a highly enriched protein with potential host insect-killing activity from the culture supernatant of P. entomophila. Then, a 45-kDa protein was purified to homogeneity and identified as AprA which is an alkaline zinc metalloprotease of the genus Pseudomonas by liquid chromatography mass spectrometry (LC-MS). Purified AprA showed a pronounced killing effect against host insects and suppressed both host cellular and humoral innate immunity. Furthermore, to show that AprA is an important insecticidal protein of P. entomophila, we used an aprA-deficient P. entomophila mutant strain (ΔaprA). When ΔaprA mutant cells were injected to host insects, this mutant exhibited extremely attenuated virulence. In addition, the cytotoxicity against host hemocytes and the antimicrobial peptide-degrading ability of the ΔaprA mutant were greatly decreased. These findings suggest that AprA functions as an important insecticidal protein of P. entomophila via suppression of host cellular and humoral innate immune responses.
Collapse
Affiliation(s)
- Seung Ah Lee
- Global Research Laboratory of Insect Symbiosis, Pusan National University, Busan 46241, South Korea
| | - Seong Han Jang
- Global Research Laboratory of Insect Symbiosis, Pusan National University, Busan 46241, South Korea
| | - Byung Hyun Kim
- Global Research Laboratory of Insect Symbiosis, Pusan National University, Busan 46241, South Korea
| | - Toshio Shibata
- Institute for Advanced Study, Kyushu University, Fukuoka 819-0395, Japan; Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Jinwook Yoo
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Shun-Ichiro Kawabata
- Institute for Advanced Study, Kyushu University, Fukuoka 819-0395, Japan; Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Bok Luel Lee
- Global Research Laboratory of Insect Symbiosis, Pusan National University, Busan 46241, South Korea.
| |
Collapse
|
102
|
Parvy JP, Hodgson JA, Cordero JB. Drosophila as a Model System to Study Nonautonomous Mechanisms Affecting Tumour Growth and Cell Death. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7152962. [PMID: 29725601 PMCID: PMC5872677 DOI: 10.1155/2018/7152962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/04/2018] [Indexed: 12/26/2022]
Abstract
The study of cancer has represented a central focus in medical research for over a century. The great complexity and constant evolution of the pathology require the use of multiple research model systems and interdisciplinary approaches. This is necessary in order to achieve a comprehensive understanding into the mechanisms driving disease initiation and progression, to aid the development of appropriate therapies. In recent decades, the fruit fly Drosophila melanogaster and its associated powerful genetic tools have become a very attractive model system to study tumour-intrinsic and non-tumour-derived processes that mediate tumour development in vivo. In this review, we will summarize recent work on Drosophila as a model system to study cancer biology. We will focus on the interactions between tumours and their microenvironment, including extrinsic mechanisms affecting tumour growth and how tumours impact systemic host physiology.
Collapse
Affiliation(s)
- Jean-Philippe Parvy
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Joseph A. Hodgson
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Julia B. Cordero
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
103
|
Abstract
The proto-oncogene Myc is well known for its roles in promoting cell growth, proliferation and apoptosis. However, in this study, we found from a genetic screen that Myc inhibits, rather than promotes, cell death triggered by c-Jun N-terminal kinase (JNK) signaling in Drosophila. Firstly, expression of Drosophila Myc (dMyc) suppresses, whereas loss of dMyc enhances, ectopically activated JNK signaling-induced cell death. Secondly, dMyc impedes physiologically activated JNK pathway-mediated cell death. Thirdly, loss of dMyc triggers JNK pathway activation and JNK-dependent cell death. Finally, the mammalian cMyc gene, when expressed in Drosophila, impedes activated JNK signaling-induced cell death. Thus, besides its well-studied apoptosis promoting function, Myc also antagonizes JNK-mediated cell death in Drosophila, and this function is likely conserved from fly to human.
Collapse
Affiliation(s)
- Jiuhong Huang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Feng
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xinhong Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Wenzhe Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
104
|
Mondal T, Bag I, SNCVL P, Garikapati KR, Bhadra U, Pal Bhadra M. Two way controls of apoptotic regulators consign DmArgonaute-1 a better clasp on it. PLoS One 2018; 13:e0190548. [PMID: 29385168 PMCID: PMC5791970 DOI: 10.1371/journal.pone.0190548] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/16/2017] [Indexed: 02/02/2023] Open
Abstract
Argonaute family proteins are well conserved among all organisms. Its role in mitotic cell cycle progression and apoptotic cell elimination is poorly understood. Earlier we have established the contribution of Ago-1 in cell cycle control related to G2/M cyclin in Drosophila. Here we have extended our study in understanding the relationship of Ago-1 in regulating apoptosis during Drosophila development. Apoptosis play a critical role in controlling organ shape and size during development of multi cellular organism. Multifarious regulatory pathways control apoptosis during development among which highly conserved JNK (c-Jun N-terminal kinase) pathway play a crucial role. Here we have over expressed Ago-1 in Drosophila eye and brain by employing UAS (upstream activation sequence)-GAL4 system under the expression of eye and brain specific driver. Over expression of Ago-1 resulted in reduced number of ommatidia in the eye and produced smaller size brain in adult and larval Drosophila. A drastic reversal of the phenotype towards normal was observed upon introduction of a single copy of the dominant negative mutation of basket (bsk, Drosophila homolog of JNK) indicating an active and physical involvement of the bsk with Ago-1 in inducing developmental apoptotic process. Further study showed that Ago-1 stimulates phosphorylation of JNK through transforming growth factor-β activated kinase 1- hemipterous (Tak1-hep) axis of JNK pathway. JNK phosphorylation results in up regulation of pro-apoptotic genes head involution defective (hid), grim & reaper (rpr) and induces activation of Drosophila caspases (cysteinyl aspartate proteinases);DRONC (Death regulator Nedd2-like caspase), ICE (alternatively Drice, Death related ICE-like caspase) and DCP1 (Death caspase-1) by inhibiting apoptotic inhibitor protein DIAP1 (Death-associated inhibitor of apoptosis 1). Further, Ago-1 also inhibits miR-14 expression to trigger apoptosis. Our findings propose that Ago-1 acts as a key regulator in controlling cell death, tumor regression and stress response in metazoan providing a constructive bridge between RNAi machinery and cell death.
Collapse
Affiliation(s)
- Tanmoy Mondal
- Department of Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana State, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India
| | - Indira Bag
- Department of Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana State, India
| | - Pushpavalli SNCVL
- Department of Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana State, India
| | - Koteswara Rao Garikapati
- Department of Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana State, India
| | - Utpal Bhadra
- Gene Silencing and Functional Genomics Group, CSIR-Centre For Cellular and Molecular Biology, Uppal Road, Hyderabad, Telangana State, India
| | - Manika Pal Bhadra
- Department of Chemical Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana State, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IICT Campus, Hyderabad, India
- * E-mail: ,
| |
Collapse
|
105
|
Jiang Q, Bao C, Yang Y, Liu A, Liu F, Huang H, Ye H. Transcriptome profiling of claw muscle of the mud crab (Scylla paramamosain) at different fattening stages. PLoS One 2017; 12:e0188067. [PMID: 29141033 PMCID: PMC5687733 DOI: 10.1371/journal.pone.0188067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/31/2017] [Indexed: 01/29/2023] Open
Abstract
In crustaceans, muscle growth and development is complicated, and to date substantial knowledge gaps exist. In this study, the claw muscle, hepatopancreas and nervous tissue of the mud crab (Scylla paramamosain) were collected at three fattening stages for sequence by the Illumina sequencing. A total of 127.87 Gb clean data with no less than 3.94 Gb generated for each sample and the cycleQ30 percentages were more than 86.13% for all samples. De Bruijn assembly of these clean data produced 94,853 unigenes, thereinto, 50,059 unigenes were found in claw muscle. A total of 121 differentially expressed genes (DEGs) were revealed in claw muscle from the three fattening stages with a Padj value < 0.01, including 63 genes with annotation. Functional annotation and enrichment analysis showed that the DEGs clusters represented the predominant gene catalog with roles in biochemical processes (glycolysis, phosphorylation and regulation of transcription), molecular function (ATP binding, 6-phosphofructokinase activity, and sequence-specific DNA binding) and cellular component (6-phosphofructokinase complex, plasma membrane, and integral component of membrane). qRT-PCR was employed to further validate certain DEGs. Single nucleotide polymorphism (SNP) analysis obtained 159,322, 125,963 and 166,279 potential SNPs from the muscle transcriptome at stage B, stage C and stage D, respectively. In addition, there were sixteen neuropeptide transcripts being predicted in the claw muscle. The present study provides a comprehensive transcriptome of claw muscle of S. paramamosain during fattening, providing a basis for screening the functional genes that may affect muscle growth of S. paramamosain.
Collapse
Affiliation(s)
- Qingling Jiang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Chenchang Bao
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ya’nan Yang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - An Liu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fang Liu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Huiyang Huang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Haihui Ye
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Collaborative Innovation Center for Development and Utilization of Marine Biological Resources, Xiamen, China
- * E-mail:
| |
Collapse
|
106
|
Chen L, Xiong YQ, Xu J, Wang JP, Meng ZL, Hong YQ. Juglanin inhibits lung cancer by regulation of apoptosis, ROS and autophagy induction. Oncotarget 2017; 8:93878-93898. [PMID: 29212196 PMCID: PMC5706842 DOI: 10.18632/oncotarget.21317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/27/2017] [Indexed: 12/31/2022] Open
Abstract
Juglanin (Jug) is obtained from the crude extract of Polygonum aviculare, exerting suppressive activity against cancer cell progression in vitro and in vivo. Juglanin administration causes apoptosis and reactive oxygen species (ROS) in different types of cells through regulating various signaling pathways. In our study, the effects of juglanin on non-small cell lung cancer were investigated. A significant role of juglanin in suppressing lung cancer growth was observed. Juglanin promoted apoptosis in lung cancer cells through increasing Caspase-3 and poly ADP-ribose polymerase (PARP) cleavage, which is regulated by TNF-related apoptosis-inducing ligand/Death receptors (TRAIL/DRs) relied on p53 activation. Anti-apoptotic members Bcl-2 and Bcl-xl were reduced, and pro-apoptotic members Bax and Bad were enhanced in cells and animals receiving juglanin. Additionally, nuclear factor-κB (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and mitogen-activated protein kinases (MAPKs) activation were inhibited by juglanin. Further, juglanin improved ROS and induced autophagy. ROS inhibitor N-acetyl-l-cysteine (NAC) reversed apoptosis induced by juglanin in cancer cells. The formation of autophagic vacoules and LC3/autophagy gene7 (ATG7)/Beclin1 (ATG6) over-expression were observed in juglanin-treated cells. Also, juglanin administration to mouse xenograft models inhibited lung cancer progression. Our study demonstrated that juglanin could be a promising candidate against human lung cancer progression.
Collapse
Affiliation(s)
- Liang Chen
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Ya-Qiong Xiong
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Jing Xu
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Ji-Peng Wang
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Zi-Li Meng
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Yong-Qing Hong
- Department of Respiration, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| |
Collapse
|
107
|
Upadhyay A, Moss-Taylor L, Kim MJ, Ghosh AC, O'Connor MB. TGF-β Family Signaling in Drosophila. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022152. [PMID: 28130362 DOI: 10.1101/cshperspect.a022152] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The transforming growth factor β (TGF-β) family signaling pathway is conserved and ubiquitous in animals. In Drosophila, fewer representatives of each signaling component are present compared with vertebrates, simplifying mechanistic study of the pathway. Although there are fewer family members, the TGF-β family pathway still regulates multiple and diverse functions in Drosophila. In this review, we focus our attention on several of the classic and best-studied functions for TGF-β family signaling in regulating Drosophila developmental processes such as embryonic and imaginal disc patterning, but we also describe several recently discovered roles in regulating hormonal, physiological, neuronal, innate immunity, and tissue homeostatic processes.
Collapse
Affiliation(s)
- Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Lindsay Moss-Taylor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Arpan C Ghosh
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
108
|
Pérez E, Lindblad JL, Bergmann A. Tumor-promoting function of apoptotic caspases by an amplification loop involving ROS, macrophages and JNK in Drosophila. eLife 2017; 6:e26747. [PMID: 28853394 PMCID: PMC5779227 DOI: 10.7554/elife.26747] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/28/2017] [Indexed: 01/04/2023] Open
Abstract
Apoptosis and its molecular mediators, the caspases, have long been regarded as tumor suppressors and one hallmark of cancer is 'Evading Apoptosis'. However, recent work has suggested that apoptotic caspases can also promote proliferation and tumor growth under certain conditions. How caspases promote proliferation and how cells are protected from the potentially harmful action of apoptotic caspases is largely unknown. Here, we show that although caspases are activated in a well-studied neoplastic tumor model in Drosophila, oncogenic mutations of the proto-oncogene Ras (RasV12) maintain tumorous cells in an 'undead'-like condition and transform caspases from tumor suppressors into tumor promotors. Instead of killing cells, caspases now promote the generation of intra- and extracellular reactive oxygen species (ROS). One function of the ROS is the recruitment and activation of macrophage-like immune cells which in turn signal back to tumorous epithelial cells to activate oncogenic JNK signaling. JNK further promotes and amplifies caspase activity, thereby constituting a feedback amplification loop. Interfering with the amplification loop strongly reduces the neoplastic behavior of these cells and significantly improves organismal survival. In conclusion, RasV12-modified caspases initiate a feedback amplification loop involving tumorous epithelial cells and macrophage-like immune cells that is necessary for uncontrolled tumor growth and invasive behavior.
Collapse
Affiliation(s)
- Ernesto Pérez
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Jillian L Lindblad
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Andreas Bergmann
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|
109
|
Abstract
While several large-scale resources are available for in vivo loss-of-function studies in Drosophila, an analogous resource for overexpressing genes from their endogenous loci does not exist. We describe a strategy for generating such a resource using Cas9 transcriptional activators (CRISPRa). First, we compare a panel of CRISPRa approaches and demonstrate that, for in vivo studies, dCas9-VPR is the most optimal activator. Next, we demonstrate that this approach is scalable and has a high success rate, as >75% of the lines tested activate their target gene. We show that CRISPRa leads to physiologically relevant levels of target gene expression capable of generating strong gain-of-function (GOF) phenotypes in multiple tissues and thus serves as a useful platform for genetic screening. Based on the success of this CRISRPa approach, we are generating a genome-wide collection of flies expressing single-guide RNAs (sgRNAs) for CRISPRa. We also present a collection of more than 30 Gal4 > UAS:dCas9-VPR lines to aid in using these sgRNA lines for GOF studies in vivo.
Collapse
|
110
|
Feedback amplification loop drives malignant growth in epithelial tissues. Proc Natl Acad Sci U S A 2017; 114:E7291-E7300. [PMID: 28808034 DOI: 10.1073/pnas.1701791114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Interactions between cells bearing oncogenic mutations and the surrounding microenvironment, and cooperation between clonally distinct cell populations, can contribute to the growth and malignancy of epithelial tumors. The genetic techniques available in Drosophila have contributed to identify important roles of the TNF-α ligand Eiger and mitogenic molecules in mediating these interactions during the early steps of tumor formation. Here we unravel the existence of a tumor-intrinsic-and microenvironment-independent-self-reinforcement mechanism that drives tumor initiation and growth in an Eiger-independent manner. This mechanism relies on cell interactions between two functionally distinct cell populations, and we present evidence that these cell populations are not necessarily genetically different. Tumor-specific and cell-autonomous activation of the tumorigenic JNK stress-activated pathway drives the expression of secreted signaling molecules and growth factors to delaminating cells, which nonautonomously promote proliferative growth of the partially transformed epithelial tissue. We present evidence that cross-feeding interactions between delaminating and nondelaminating cells increase each other's sizes and that these interactions can explain the unlimited growth potential of these tumors. Our results will open avenues toward our molecular understanding of those social cell interactions with a relevant function in tumor initiation in humans.
Collapse
|
111
|
Abstract
Cell death is a fundamental progress that regulates cell number, tissue homeostasis and organ size in development. The c-Jun N-terminal kinase (JNK) pathway has been evolutionarily conserved from fly to human, and plays essential roles in regulating cell death. To characterize additional genes that regulate JNK signaling, we performed a genetic screen in Drosophila and identified dGLYAT, a novel gene whose function was previously unknown, as a modulator of JNK-mediated cell death. We found that loss of dGLYAT suppressed JNK activation and cell death triggered by over-expression of Egr or Hep, or depletion of puc or lgl in development, suggesting dGLYAT regulates both ectopic and physiological functions of JNK pathway. Furthermore, we showed that loss of dGLYAT inhibits JNK-mediated ROS production, suggesting dGLYAT regulates multiple functions of JNK signaling in vivo.
Collapse
|
112
|
Vaughen J, Igaki T. Slit-Robo Repulsive Signaling Extrudes Tumorigenic Cells from Epithelia. Dev Cell 2017; 39:683-695. [PMID: 27997825 DOI: 10.1016/j.devcel.2016.11.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/04/2016] [Accepted: 11/19/2016] [Indexed: 02/08/2023]
Abstract
Cells dynamically interact throughout animal development to coordinate growth and deter disease. For example, cell-cell competition weeds out aberrant cells to enforce homeostasis. In Drosophila, tumorigenic cells mutant for the cell polarity gene scribble (scrib) are actively eliminated from epithelia when surrounded by wild-type cells. While scrib cell elimination depends critically on JNK signaling, JNK-dependent cell death cannot sufficiently explain scrib cell extirpation. Thus, how JNK executed cell elimination remained elusive. Here, we show that repulsive Slit-Robo2-Ena signaling exerts an extrusive force downstream of JNK to eliminate scrib cells from epithelia by disrupting E-cadherin. While loss of Slit-Robo2-Ena in scrib cells potentiates scrib tumor formation within the epithelium, Robo2-Ena hyperactivation surprisingly triggers luminal scrib tumor growth following excess extrusion. This extrusive signaling is amplified by a positive feedback loop between Slit-Robo2-Ena and JNK. Our observations provide a potential causal mechanism for Slit-Robo dysregulation in numerous human cancers.
Collapse
Affiliation(s)
- John Vaughen
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
113
|
Jo J, Im SH, Babcock DT, Iyer SC, Gunawan F, Cox DN, Galko MJ. Drosophila caspase activity is required independently of apoptosis to produce active TNF/Eiger during nociceptive sensitization. Cell Death Dis 2017; 8:e2786. [PMID: 28492538 PMCID: PMC5520682 DOI: 10.1038/cddis.2016.474] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/03/2016] [Accepted: 12/06/2016] [Indexed: 01/12/2023]
Abstract
Tumor necrosis factor (TNF) signaling is required for inflammatory nociceptive (pain) sensitization in Drosophila and vertebrates. Nociceptive sensitization in Drosophila larvae following UV-induced tissue damage is accompanied by epidermal apoptosis and requires epidermal-derived TNF/Eiger and the initiator caspase, Dronc. Major gaps remain regarding TNF function in sensitization, including the relationship between apoptosis/tissue damage and TNF production, the downstream signaling in this context, and the target genes that modulate nociceptive behaviors. Here, apoptotic cell death and thermal nociceptive sensitization are genetically and procedurally separable in a Drosophila model of UV-induced nociceptive sensitization. Activation of epidermal Dronc induces TNF-dependent but effector caspase-independent nociceptive sensitization in the absence of UV. In addition, knockdown of Dronc attenuated nociceptive sensitization induced by full-length TNF/Eiger but not by a constitutively soluble form. UV irradiation induced TNF production in both in vitro and in vivo, but TNF secretion into hemolymph was not sufficient to induce thermal nociceptive sensitization. Downstream mediators of TNF-induced sensitization included two TNF receptor-associated factors, a p38 kinase, and the transcription factor nuclear factor kappa B. Finally, sensory neuron-specific microarray analysis revealed downstream TNF target genes induced during thermal nociceptive sensitization. One of these, enhancer of zeste (E(z)), functions downstream of TNF during thermal nociceptive sensitization. Our findings suggest that an initiator caspase is involved in TNF processing/secretion during nociceptive sensitization, and that TNF activation leads to a specific downstream signaling cascade and gene transcription required for sensitization. These findings have implications for both the evolution of inflammatory caspase function following tissue damage signals and the action of TNF during sensitization in vertebrates.
Collapse
Affiliation(s)
- Juyeon Jo
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Genes and Development Graduate Program, Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Seol Hee Im
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel T Babcock
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Srividya C Iyer
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Felona Gunawan
- Department of Biochemistry and Cell Biology, Rice University, Houston, TX, USA
| | - Daniel N Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Michael J Galko
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Genes and Development Graduate Program, Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
114
|
Lee JH, Lee CW, Park SH, Choe KM. Spatiotemporal regulation of cell fusion by JNK and JAK/STAT signaling during Drosophila wound healing. J Cell Sci 2017; 130:1917-1928. [PMID: 28424232 DOI: 10.1242/jcs.187658] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 04/18/2017] [Indexed: 12/23/2022] Open
Abstract
Cell-cell fusion is widely observed during development and disease, and imposes a dramatic change on participating cells. Cell fusion should be tightly controlled, but the underlying mechanism is poorly understood. Here, we found that the JAK/STAT pathway suppressed cell fusion during wound healing in the Drosophila larval epidermis, restricting cell fusion to the vicinity of the wound. In the absence of JAK/STAT signaling, a large syncytium containing a 3-fold higher number of nuclei than observed in wild-type tissue formed in wounded epidermis. The JAK/STAT ligand-encoding genes upd2 and upd3 were transcriptionally induced by wounding, and were required for suppressing excess cell fusion. JNK (also known as Basket in flies) was activated in the wound vicinity and activity peaked at ∼8 h after injury, whereas JAK/STAT signaling was activated in an adjoining concentric ring and activity peaked at a later stage. Cell fusion occurred primarily in the wound vicinity, where JAK/STAT activation was suppressed by fusion-inducing JNK signaling. JAK/STAT signaling was both necessary and sufficient for the induction of βPS integrin (also known as Myospheroid) expression, suggesting that the suppression of cell fusion was mediated at least in part by integrin protein.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Chan-Wool Lee
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Si-Hyoung Park
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Kwang-Min Choe
- Department of Systems Biology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| |
Collapse
|
115
|
Killers creating new life: caspases drive apoptosis-induced proliferation in tissue repair and disease. Cell Death Differ 2017; 24:1390-1400. [PMID: 28362431 PMCID: PMC5520457 DOI: 10.1038/cdd.2017.47] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/26/2017] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
Apoptosis is a carefully orchestrated and tightly controlled form of cell death, conserved across metazoans. As the executioners of apoptotic cell death, cysteine-dependent aspartate-directed proteases (caspases) are critical drivers of this cellular disassembly. Early studies of genetically programmed cell death demonstrated that the selective activation of caspases induces apoptosis and the precise elimination of excess cells, thereby sculpting structures and refining tissues. However, over the past decade there has been a fundamental shift in our understanding of the roles of caspases during cell death-a shift precipitated by the revelation that apoptotic cells actively engage with their surrounding environment throughout the death process, and caspases can trigger a myriad of signals, some of which drive concurrent cell proliferation regenerating damaged structures and building up lost tissues. This caspase-driven compensatory proliferation is referred to as apoptosis-induced proliferation (AiP). Diverse mechanisms of AiP have been found across species, ranging from planaria to mammals. In this review, we summarize the current knowledge of AiP and we highlight recent advances in the field including the involvement of reactive oxygen species and macrophage-like immune cells in one form of AiP, novel regulatory mechanisms affecting caspases during AiP, and emerging clinical data demonstrating the critical importance of AiP in cancer.
Collapse
|
116
|
Casas-Tintó S, Arnés M, Ferrús A. Drosophila enhancer-Gal4 lines show ectopic expression during development. ROYAL SOCIETY OPEN SCIENCE 2017; 4:170039. [PMID: 28405401 PMCID: PMC5383858 DOI: 10.1098/rsos.170039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/27/2017] [Indexed: 06/07/2023]
Abstract
In Drosophila melanogaster the most widely used technique to drive gene expression is the binary UAS/Gal4 system. We show here that a set of nervous system specific enhancers (elav, D42/Toll-6, OK6/RapGAP1) display ectopic activity in epithelial tissues during development, which is seldom considered in experimental studies. This ectopic activity is variable, unstable and influenced by the primary sequence of the enhancer and the insertion site in the chromosome. In addition, the ectopic activity is independent of the protein expressed, Gal4, as it is reproduced also with the expression of Gal80. Another enhancer, LN2 from the sex lethal (Sxl) gene, shows sex-dependent features in its ectopic expression. Feminization of LN2 expressing males does not alter the male specific pattern indicating that the sexual dimorphism of LN2 expression is an intrinsic feature of this enhancer. Other X chromosome enhancers corresponding to genes not related to sex determination do not show sexual dimorphism in their ectopic expressions. Although variable and unstable, the ectopic activation of enhancer-Gal4 lines seems to be regulated in terms of tissue and intensity. To characterize the full domain of expression of enhancer-Gal4 constructs is relevant for the design of transgenic animal models and biotechnology tools, as well as for the correct interpretation of developmental and behavioural studies in which Gal4 lines are used.
Collapse
|
117
|
Diaz-Garcia S, Ahmed S, Baonza A. Analysis of the Function of Apoptosis during Imaginal Wing Disc Regeneration in Drosophila melanogaster. PLoS One 2016; 11:e0165554. [PMID: 27893747 PMCID: PMC5125585 DOI: 10.1371/journal.pone.0165554] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/13/2016] [Indexed: 01/02/2023] Open
Abstract
Regeneration is the ability that allows organisms to replace missing organs or lost tissue after injuries. This ability requires the coordinated activity of different cellular processes, including programmed cell death. Apoptosis plays a key role as a source of signals necessary for regeneration in different organisms. The imaginal discs of Drosophila melanogaster provide a particularly well-characterised model system for studying the cellular and molecular mechanisms underlying regeneration. Although it has been shown that signals produced by apoptotic cells are needed for homeostasis and regeneration of some tissues of this organism, such as the adult midgut, the contribution of apoptosis to disc regeneration remains unclear. Using a new method for studying disc regeneration in physiological conditions, we have defined the pattern of cell death in regenerating discs. Our data indicate that during disc regeneration, cell death increases first at the wound edge, but as regeneration progresses dead cells can be observed in regions far away from the site of damage. This result indicates that apoptotic signals initiated in the wound spread throughout the disc. We also present results which suggest that the partial inhibition of apoptosis does not have a major effect on disc regeneration. Finally, our results suggest that during disc regeneration distinct apoptotic signals might be acting simultaneously.
Collapse
Affiliation(s)
- Sandra Diaz-Garcia
- University of California San Diego, Biology Section of Cell & Developmental Biology, United States of America
| | - Sara Ahmed
- Centro de Biología Molecular “Severo Ochoa”, CSIC and Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Baonza
- Centro de Biología Molecular “Severo Ochoa”, CSIC and Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
118
|
Liu D, Shaukat Z, Saint RB, Gregory SL. Chromosomal instability triggers cell death via local signalling through the innate immune receptor Toll. Oncotarget 2016; 6:38552-65. [PMID: 26462024 PMCID: PMC4770720 DOI: 10.18632/oncotarget.6035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/08/2015] [Indexed: 01/29/2023] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and has been implicated in cancer initiation, progression and the development of resistance to traditional cancer therapy. Here we identify a new property of CIN cells, showing that inducing CIN in proliferating Drosophila larval tissue leads to the activation of innate immune signalling in CIN cells. Manipulation of this immune pathway strongly affects the survival of CIN cells, primarily via JNK, which responds to both Toll and TNFα/Eiger. This pathway also activates Mmp1, which recruits hemocytes to the CIN tissue to provide local amplification of the immune response that is needed for effective elimination of CIN cells.
Collapse
Affiliation(s)
- Dawei Liu
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Zeeshan Shaukat
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Robert B Saint
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Stephen L Gregory
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
119
|
The transcription factor Ets21C drives tumor growth by cooperating with AP-1. Sci Rep 2016; 6:34725. [PMID: 27713480 PMCID: PMC5054425 DOI: 10.1038/srep34725] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/08/2016] [Indexed: 01/25/2023] Open
Abstract
Tumorigenesis is driven by genetic alterations that perturb the signaling networks regulating proliferation or cell death. In order to block tumor growth, one has to precisely know how these signaling pathways function and interplay. Here, we identified the transcription factor Ets21C as a pivotal regulator of tumor growth and propose a new model of how Ets21C could affect this process. We demonstrate that a depletion of Ets21C strongly suppressed tumor growth while ectopic expression of Ets21C further increased tumor size. We confirm that Ets21C expression is regulated by the JNK pathway and show that Ets21C acts via a positive feed-forward mechanism to induce a specific set of target genes that is critical for tumor growth. These genes are known downstream targets of the JNK pathway and we demonstrate that their expression not only depends on the transcription factor AP-1, but also on Ets21C suggesting a cooperative transcriptional activation mechanism. Taken together we show that Ets21C is a crucial player in regulating the transcriptional program of the JNK pathway and enhances our understanding of the mechanisms that govern neoplastic growth.
Collapse
|
120
|
Abstract
The prognosis of patients with classical Hodgkin lymphoma following chemo- and radiotherapy has been excellent during the last 4 decades. However, the development of secondary malignancies is of major concern. Therefore, the reduction of radiotherapy application is a major objective of ongoing clinical trials. De-escalation of treatment may increase the risk of relapses and thus may lead to reappearance of prognostic factors. Prognostic biomarkers might help to identify patients who are at increased risk of relapse. This review summarizes the current knowledge about potential prognostic biomarkers for patients with classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Martin S Staege
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Stefanie Kewitz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Toralf Bernig
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Caspar Kühnöl
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Christine Mauz-Körholz
- a Department of Pediatrics , Martin Luther University Halle-Wittenberg , Halle , Germany
| |
Collapse
|
121
|
Hou L, Liu K, Li Y, Ma S, Ji X, Liu L. Necrotic pyknosis is a morphologically and biochemically distinct event from apoptotic pyknosis. J Cell Sci 2016; 129:3084-90. [PMID: 27358477 DOI: 10.1242/jcs.184374] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/23/2016] [Indexed: 12/17/2022] Open
Abstract
Classification of apoptosis and necrosis by morphological differences has been widely used for decades. However, this usefulness of this method has been seriously questioned in recent years, mainly due to a lack of functional and biochemical evidence to interpret the morphology changes. To address this matter, we devised genetic manipulations in Drosophila to study pyknosis, a process of nuclear shrinkage and chromatin condensation that occurs in apoptosis and necrosis. By following the progression of necrotic pyknosis, we surprisingly observed a transient state of chromatin detachment from the nuclear envelope, followed by the nuclear envelope completely collapsing onto chromatin. This phenomenon led us to discover that phosphorylation of barrier-to-autointegration factor (BAF) mediates this initial separation of nuclear envelope from chromatin. Functionally, inhibition of BAF phosphorylation suppressed necrosis in both Drosophila and human cells, suggesting that necrotic pyknosis is conserved in the propagation of necrosis. In contrast, during apoptotic pyknosis the chromatin did not detach from the nuclear envelope and inhibition of BAF phosphorylation had no effect on apoptotic pyknosis and apoptosis. Our research provides the first genetic evidence supporting a morphological classification of apoptosis and necrosis through different forms of pyknosis.
Collapse
Affiliation(s)
- Lin Hou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Kai Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yuhong Li
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Shuang Ma
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Xunming Ji
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| | - Lei Liu
- Aging and Disease Laboratory of Xuanwu Hospital and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Youanmen, Beijing 100069, China
| |
Collapse
|
122
|
Eroglu M, Derry WB. Your neighbours matter - non-autonomous control of apoptosis in development and disease. Cell Death Differ 2016; 23:1110-8. [PMID: 27177021 PMCID: PMC4946894 DOI: 10.1038/cdd.2016.41] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/14/2016] [Accepted: 04/07/2016] [Indexed: 12/15/2022] Open
Abstract
Traditionally, the regulation of apoptosis has been thought of as an autonomous process in which the dying cell dictates its own demise. However, emerging studies in genetically tractable multicellular organisms, such as Caenorhabditis elegans and Drosophila, have revealed that death is often a communal event. Here, we review the current literature on non-autonomous mechanisms governing apoptosis in multiple cellular contexts. The importance of the cellular community in dictating the funeral arrangements of apoptotic cells has profound implications in development and disease.
Collapse
Affiliation(s)
- M Eroglu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - W B Derry
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
123
|
Xing Q, Yu Q, Dou H, Wang J, Li R, Ning X, Wang R, Wang S, Zhang L, Hu X, Bao Z. Genome-wide identification, characterization and expression analyses of two TNFRs in Yesso scallop (Patinopecten yessoensis) provide insight into the disparity of responses to bacterial infections and heat stress in bivalves. FISH & SHELLFISH IMMUNOLOGY 2016; 52:44-56. [PMID: 26988286 DOI: 10.1016/j.fsi.2016.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/28/2016] [Accepted: 03/10/2016] [Indexed: 05/16/2023]
Abstract
Tumor necrosis factors receptors (TNFRs) comprise a superfamily of proteins characterized by a unique cysteine-rich domain (CRD) and play important roles in diverse physiological and pathological processes in the innate immune system, including inflammation, apoptosis, autoimmunity and organogenesis. Although significant effects of TNFRs on immunity have been reported in most vertebrates as well as some invertebrates, the complete TNFR superfamily has not been systematically characterized in scallops. In this study, two different types of TNFR-like genes, including PyTNFR1 and PyTNFR2 genes were identified from Yesso scallop (Patinopecten yessoensis, Jay, 1857) through whole-genome scanning. Phylogenetic and protein structural analyses were carried out to determine the identities and evolutionary relationships of the two genes. The expression profiling of PyTNFRs was performed at different development stages, in healthy adult tissues and in hemocytes after bacterial infection and heat stress. Expression analysis revealed that both PyTNFRs were significantly induced during the acute phase (3 h) after infection with Gram-positive (Micrococcus luteus) and Gram-negative (Vibrio anguillarum) bacteria, though much more dramatic chronic-phase (24 h) changes were observed after V. anguillarum challenge. For heat stress, only PyTNFR2 displayed significant elevation at 12 h and 24 h, which suggests a functional difference in the two PyTNFRs. Collectively, this study provides novel insight into the PyTNFRs and the specific role and response of TNFR-involved pathways in host immune responses against different bacterial pathogens and heat stress in bivalves.
Collapse
Affiliation(s)
- Qiang Xing
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Qian Yu
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Huaiqian Dou
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jing Wang
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ruojiao Li
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xianhui Ning
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Ruijia Wang
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Shi Wang
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Lingling Zhang
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Xiaoli Hu
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Zhenmin Bao
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| |
Collapse
|
124
|
Agrawal N, Delanoue R, Mauri A, Basco D, Pasco M, Thorens B, Léopold P. The Drosophila TNF Eiger Is an Adipokine that Acts on Insulin-Producing Cells to Mediate Nutrient Response. Cell Metab 2016; 23:675-84. [PMID: 27076079 DOI: 10.1016/j.cmet.2016.03.003] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/25/2015] [Accepted: 03/11/2016] [Indexed: 12/27/2022]
Abstract
Adaptation of organisms to ever-changing nutritional environments relies on sensor tissues and systemic signals. Identification of these signals would help understand the physiological crosstalk between organs contributing to growth and metabolic homeostasis. Here we show that Eiger, the Drosophila TNF-α, is a metabolic hormone that mediates nutrient response by remotely acting on insulin-producing cells (IPCs). In the condition of nutrient shortage, a metalloprotease of the TNF-α converting enzyme (TACE) family is active in fat body (adipose-like) cells, allowing the cleavage and release of adipose Eiger in the hemolymph. In the brain IPCs, Eiger activates its receptor Grindelwald, leading to JNK-dependent inhibition of insulin production. Therefore, we have identified a humoral connexion between the fat body and the brain insulin-producing cells relying on TNF-α that mediates adaptive response to nutrient deprivation.
Collapse
Affiliation(s)
- Neha Agrawal
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Renald Delanoue
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Alessandra Mauri
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Davide Basco
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Matthieu Pasco
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland
| | - Pierre Léopold
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France.
| |
Collapse
|
125
|
Ruan W, Srinivasan A, Lin S, Kara KI, Barker PA. Eiger-induced cell death relies on Rac1-dependent endocytosis. Cell Death Dis 2016; 7:e2181. [PMID: 27054336 PMCID: PMC4855659 DOI: 10.1038/cddis.2016.80] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/02/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022]
Abstract
Signaling via tumor necrosis factor receptor (TNFR) superfamily members regulates cellular life and death decisions. A subset of mammalian TNFR proteins, most notably the p75 neurotrophin receptor (p75NTR), induces cell death through a pathway that requires activation of c-Jun N-terminal kinases (JNKs). However the receptor-proximal signaling events that mediate this remain unclear. Drosophila express a single tumor necrosis factor (TNF) ligand termed Eiger (Egr) that activates JNK-dependent cell death. We have exploited this model to identify phylogenetically conserved signaling events that allow Egr to induce JNK activation and cell death in vivo. Here we report that Rac1, a small GTPase, is specifically required in Egr-mediated cell death. rac1 loss of function blocks Egr-induced cell death, whereas Rac1 overexpression enhances Egr-induced killing. We identify Vav as a GEF for Rac1 in this pathway and demonstrate that dLRRK functions as a negative regulator of Rac1 that normally acts to constrain Egr-induced death. Thus dLRRK loss of function increases Egr-induced cell death in the fly. We further show that Rac1-dependent entry of Egr into early endosomes is a crucial prerequisite for JNK activation and for cell death and show that this entry requires the activity of Rab21 and Rab7. These findings reveal novel regulatory mechanisms that allow Rac1 to contribute to Egr-induced JNK activation and cell death.
Collapse
Affiliation(s)
- W Ruan
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - A Srinivasan
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - S Lin
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - k-I Kara
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - P A Barker
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.,Department of Biology, The University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| |
Collapse
|
126
|
Pozzolini M, Scarfì S, Ghignone S, Mussino F, Vezzulli L, Cerrano C, Giovine M. Molecular characterization and expression analysis of the first Porifera tumor necrosis factor superfamily member and of its putative receptor in the marine sponge Chondrosia reniformis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 57:88-98. [PMID: 26705701 DOI: 10.1016/j.dci.2015.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 12/14/2015] [Accepted: 12/14/2015] [Indexed: 06/05/2023]
Abstract
Here we report the molecular cloning and characterization of the first Tumor Necrosis Factor homologous and of its putative receptor in the marine sponge Chondrosia reniformis: chTNF and chTNFR, respectively. The deduced chTNF amino acid sequence is a type II transmembrane protein containing the typical TNFSF domain. Phylogenetic analysis reveals that chTNF is more related to Chordata TNFs rather than to other invertebrates. chTNF and chTNFR are constitutively expressed both in the ectosome and in the choanosome of the sponge, with higher levels in the ectosome. chTNF and chTNFR mRNAs were monitored in sponge fragmorphs treated with Gram(+) or Gram(-) bacteria. chTNF was significantly upregulated in Gram(+)-treated fragmorphs as compared to controls, while chTNFR was upregulated by both treatments. Finally, the possible chTNF fibrogenic role in sponge fragmorphs was studied by TNF inhibitor treatment measuring fibrillar and non fibrillar collagen gene expression; results indicate that the cytokine is involved in sponge collagen deposition and homeostasis.
Collapse
Affiliation(s)
- Marina Pozzolini
- Department of Earth, Environment and Life Sciences (DiSTAV), University of Genova, Via Pastore 3, 16132, Italy.
| | - Sonia Scarfì
- Department of Earth, Environment and Life Sciences (DiSTAV), University of Genova, Via Pastore 3, 16132, Italy
| | - Stefano Ghignone
- Institute for Sustainable Plant Protection-Turin Unit (CNR), Viale Mattioli 25, 10125 Torino, Italy
| | - Francesca Mussino
- Department of Earth, Environment and Life Sciences (DiSTAV), University of Genova, Via Pastore 3, 16132, Italy
| | - Luigi Vezzulli
- Department of Earth, Environment and Life Sciences (DiSTAV), University of Genova, Via Pastore 3, 16132, Italy
| | - Carlo Cerrano
- Department of Life and Environment Sciences (DiSVA), Marche Polytechnic University, Via Brecce Bianche, 60131 Ancona, Italy
| | - Marco Giovine
- Department of Earth, Environment and Life Sciences (DiSTAV), University of Genova, Via Pastore 3, 16132, Italy
| |
Collapse
|
127
|
Wu C, Chen C, Dai J, Zhang F, Chen Y, Li W, Pastor-Pareja JC, Xue L. Toll pathway modulates TNF-induced JNK-dependent cell death in Drosophila. Open Biol 2016. [PMID: 26202785 PMCID: PMC4632500 DOI: 10.1098/rsob.140171] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Signalling networks that control the life or death of a cell are of central interest in modern biology. While the defined roles of the c-Jun N-terminal kinase (JNK) pathway in regulating cell death have been well-established, additional factors that modulate JNK-mediated cell death have yet to be fully elucidated. To identify novel regulators of JNK-dependent cell death, we performed a dominant-modifier screen in Drosophila and found that the Toll pathway participates in JNK-mediated cell death. Loss of Toll signalling suppresses ectopically and physiologically activated JNK signalling-induced cell death. Our epistasis analysis suggests that the Toll pathway acts as a downstream modulator for JNK-dependent cell death. In addition, gain of JNK signalling results in Toll pathway activation, revealed by stimulated transcription of Drosomycin (Drs) and increased cytoplasm-to-nucleus translocation of Dorsal. Furthermore, the Spätzle (Spz) family ligands for the Toll receptor are transcriptionally upregulated by activated JNK signalling in a non-cell-autonomous manner, providing a molecular mechanism for JNK-induced Toll pathway activation. Finally, gain of Toll signalling exacerbates JNK-mediated cell death and promotes cell death independent of caspases. Thus, we have identified another important function for the evolutionarily conserved Toll pathway, in addition to its well-studied roles in embryonic dorso-ventral patterning and innate immunity.
Collapse
Affiliation(s)
- Chenxi Wu
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| | - Changyan Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| | - Jianli Dai
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, People's Republic of China
| | - Fan Zhang
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| | - Yujun Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| | - Wenzhe Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, People's Republic of China
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, People's Republic of China
| |
Collapse
|
128
|
Extracellular Reactive Oxygen Species Drive Apoptosis-Induced Proliferation via Drosophila Macrophages. Curr Biol 2016; 26:575-84. [PMID: 26898463 DOI: 10.1016/j.cub.2015.12.064] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 11/16/2015] [Accepted: 12/16/2015] [Indexed: 12/22/2022]
Abstract
Apoptosis-induced proliferation (AiP) is a compensatory mechanism to maintain tissue size and morphology following unexpected cell loss during normal development, and may also be a contributing factor to cancer and drug resistance. In apoptotic cells, caspase-initiated signaling cascades lead to the downstream production of mitogenic factors and the proliferation of neighboring surviving cells. In epithelial cells of Drosophila imaginal discs, the Caspase-9 ortholog Dronc drives AiP via activation of Jun N-terminal kinase (JNK); however, the specific mechanisms of JNK activation remain unknown. Here we show that caspase-induced activation of JNK during AiP depends on an inflammatory response. This is mediated by extracellular reactive oxygen species (ROSs) generated by the NADPH oxidase Duox in epithelial disc cells. Extracellular ROSs activate Drosophila macrophages (hemocytes), which in turn trigger JNK activity in epithelial cells by signaling through the tumor necrosis factor (TNF) ortholog Eiger. We propose that in an immortalized ("undead") model of AiP, signaling back and forth between epithelial disc cells and hemocytes by extracellular ROSs and TNF/Eiger drives overgrowth of the disc epithelium. These data illustrate a bidirectional cell-cell communication pathway with implication for tissue repair, regeneration, and cancer.
Collapse
|
129
|
Yasin HWR, van Rensburg SH, Feiler CE, Johnson RI. The adaptor protein Cindr regulates JNK activity to maintain epithelial sheet integrity. Dev Biol 2016; 410:135-149. [PMID: 26772997 DOI: 10.1016/j.ydbio.2016.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/23/2015] [Accepted: 01/04/2016] [Indexed: 12/19/2022]
Abstract
Epithelia are essential barrier tissues that must be appropriately maintained for their correct function. To achieve this a plethora of protein interactions regulate epithelial cell number, structure and adhesion, and differentiation. Here we show that Cindr (the Drosophila Cin85 and Cd2ap ortholog) is required to maintain epithelial integrity. Reducing Cindr triggered cell delamination and movement. Most delaminating cells died. These behaviors were consistent with JNK activation previously associated with loss of epithelial integrity in response to ectopic oncogene activity. We confirmed a novel interaction between Cindr and Drosophila JNK (dJNK), which when perturbed caused inappropriate JNK signaling. Genetically reducing JNK signaling activity suppressed the effects of reducing Cindr. Furthermore, ectopic JNK signaling phenocopied loss of Cindr and was partially rescued by concomitant cindr over-expression. Thus, correct Cindr-dJNK stoichiometry is essential to maintain epithelial integrity and disturbing this balance may contribute to the pathogenesis of disease states, including cancer.
Collapse
Affiliation(s)
- Hannah W R Yasin
- Biology Department, Wesleyan University, 52 Lawn Avenue, Middletown, CT, USA
| | | | - Christina E Feiler
- Biology Department, Wesleyan University, 52 Lawn Avenue, Middletown, CT, USA
| | - Ruth I Johnson
- Biology Department, Wesleyan University, 52 Lawn Avenue, Middletown, CT, USA.
| |
Collapse
|
130
|
Apoptotic Caspases in Promoting Cancer: Implications from Their Roles in Development and Tissue Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 930:89-112. [PMID: 27558818 DOI: 10.1007/978-3-319-39406-0_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis, a major form of programmed cell death, is an important mechanism to remove extra or unwanted cells during development. In tissue homeostasis apoptosis also acts as a monitoring machinery to eliminate damaged cells in response to environmental stresses. During these processes, caspases, a group of proteases, have been well defined as key drivers of cell death. However, a wealth of evidence is emerging which supports the existence of many other non-apoptotic functions of these caspases, which are essential not only in proper organism development but also in tissue homeostasis and post-injury recovery. In particular, apoptotic caspases in stress-induced dying cells can activate mitogenic signals leading to proliferation of neighbouring cells, a phenomenon termed apoptosis-induced proliferation. Apparently, such non-apoptotic functions of caspases need to be controlled and restrained in a context-dependent manner during development to prevent their detrimental effects. Intriguingly, accumulating studies suggest that cancer cells are able to utilise these functions of caspases to their advantage to enable their survival, proliferation and metastasis in order to grow and progress. This book chapter will review non-apoptotic functions of the caspases in development and tissue homeostasis with focus on how these cellular processes can be hijacked by cancer cells and contribute to tumourigenesis.
Collapse
|
131
|
La Fortezza M, Schenk M, Cosolo A, Kolybaba A, Grass I, Classen AK. JAK/STAT signalling mediates cell survival in response to tissue stress. Development 2016; 143:2907-19. [DOI: 10.1242/dev.132340] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Tissue homeostasis relies on the ability of tissues to respond to stress. Tissue regeneration and tumour models in Drosophila have shown that JNK is a prominent stress-response pathway promoting injury-induced apoptosis and compensatory proliferation. A central question remaining unanswered is how both responses are balanced by activation of a single pathway. JAK/STAT signalling, a potential JNK target, is implicated in promoting compensatory proliferation. While we observe JAK/STAT activation in imaginal discs upon damage, our data demonstrates that JAK/STAT and its downstream effector Zfh2 promote survival of JNK-signalling cells instead. The JNK component fos and the pro-apoptotic gene hid are regulated in a JAK/STAT-dependent manner. This molecular pathway restrains JNK-induced apoptosis and spatial propagation of JNK-signalling, thereby limiting the extent of tissue damage, as well as facilitating systemic and proliferative responses to injury. We find that the pro-survival function of JAK/STAT also drives tumour growth under conditions of chronic stress. Our study defines JAK/STAT function in tissue stress and illustrates how crosstalk between conserved signalling pathways establishes an intricate equilibrium between proliferation, apoptosis and survival to restore tissue homeostasis.
Collapse
Affiliation(s)
- Marco La Fortezza
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Madlin Schenk
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Andrea Cosolo
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Addie Kolybaba
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Isabelle Grass
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| | - Anne-Kathrin Classen
- Ludwig-Maximilians-University Munich, Faculty of Biology, Grosshaderner Strasse 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
132
|
Vanha-Aho LM, Valanne S, Rämet M. Cytokines in Drosophila immunity. Immunol Lett 2015; 170:42-51. [PMID: 26730849 DOI: 10.1016/j.imlet.2015.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
Cytokines are a large and diverse group of small proteins that can affect many biological processes, but most commonly cytokines are known as mediators of the immune response. In the event of an infection, cytokines are produced in response to an immune stimulus, and they function as key regulators of the immune response. Cytokines come in many shapes and sizes, and although they vary greatly in structure, their functions have been well conserved in evolution. The immune signaling pathways that respond to cytokines are remarkably conserved from fly to man. Therefore, Drosophila melanogaster, provides an excellent platform for studying the biology and function of cytokines. In this review, we will describe the cytokines and cytokine-like molecules found in the fly and discuss their roles in host immunity.
Collapse
Affiliation(s)
- Leena-Maija Vanha-Aho
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland.
| | - Susanna Valanne
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech, 33014 University of Tampere, Finland; PEDEGO Research Unit, and Medical Research Center Oulu, University of Oulu and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
133
|
Zhang G, Li L, Meng J, Qi H, Qu T, Xu F, Zhang L. Molecular Basis for Adaptation of Oysters to Stressful Marine Intertidal Environments. Annu Rev Anim Biosci 2015; 4:357-81. [PMID: 26515272 DOI: 10.1146/annurev-animal-022114-110903] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Oysters that occupy estuarine and intertidal habitats have well-developed stress tolerance mechanisms to tolerate harsh and dynamically changing environments. In this review, we summarize common pathways and genomic features in oyster that are responsive to environmental stressors such as temperature, salinity, hypoxia, air exposure, pathogens, and anthropogenic pollutions. We first introduce the key genes involved in several pathways, which constitute the molecular basis for adaptation to stress. We use genome analysis to highlight the strong cellular homeostasis system, a unique adaptive characteristic of oysters. Next, we provide a global view of features of the oyster genome that contribute to stress adaptation, including oyster-specific gene expansion, highly inducible expression, and functional divergence. Finally, we review the consequences of interactions between oysters and the environment from ecological and evolutionary perspectives by discussing mass mortality and adaptive divergence among populations and related species of the genus Crassostrea. We conclude with prospects for future study.
Collapse
Affiliation(s)
- Guofan Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Li Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Jie Meng
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Haigang Qi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Tao Qu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Fei Xu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| | - Linlin Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, 266071 China;
| |
Collapse
|
134
|
Loss of flfl Triggers JNK-Dependent Cell Death in Drosophila. BIOMED RESEARCH INTERNATIONAL 2015; 2015:623573. [PMID: 26583122 PMCID: PMC4637051 DOI: 10.1155/2015/623573] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/05/2015] [Indexed: 01/21/2023]
Abstract
falafel (flfl) encodes a Drosophila homolog of human SMEK whose in vivo functions remain elusive. In this study, we performed gain-of-function and loss-of-function analysis in Drosophila and identified flfl as a negative regulator of JNK pathway-mediated cell death. While ectopic expression of flfl suppresses TNF-triggered JNK-dependent cell death, loss of flfl promotes JNK activation and cell death in the developing eye and wing. These data report for the first time an essential physiological function of flfl in maintaining tissue homeostasis and organ development. As the JNK signaling pathway has been evolutionary conserved from fly to human, a similar role of PP4R3 in JNK-mediated physiological process is speculated.
Collapse
|
135
|
Abstract
Cell death and inflammation are ancient processes of fundamental biological importance in both normal physiology and human disease pathologies. The recent observation that apoptosis regulatory components have dual roles in cell death and inflammation suggests that these proteins function, not primarily to kill, but to coordinate tissue repair and remodeling. This perspective unifies cell death components as positive regulators of tissue repair that replaces malfunctioning or damaged tissues and enhances the resilience of epithelia to insult. It is now recognized that cells that die by apoptosis do not do so silently, but release a variety of paracrine signals to communicate with their cellular environment to ensure tissue regeneration, and wound healing. Moreover, inflammatory signaling pathways, such as those emanating from the TNF receptor or Toll-related receptors, take part in cell competition to eliminate developmentally aberrant clones. Ubiquitylation has emerged as crucial mediator of signal transduction in cell death and inflammation. Here, we focus on recent advances on ubiquitin-mediated regulation of cell death and inflammation, and how this is used to regulate the defense of homeostasis.
Collapse
|
136
|
O’Keefe LV, Lee CS, Choo A, Richards RI. Tumor Suppressor WWOX Contributes to the Elimination of Tumorigenic Cells in Drosophila melanogaster. PLoS One 2015; 10:e0136356. [PMID: 26302329 PMCID: PMC4547717 DOI: 10.1371/journal.pone.0136356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/31/2015] [Indexed: 01/27/2023] Open
Abstract
WWOX is a >1Mb gene spanning FRA16D Common Chromosomal Fragile Site, a region of DNA instability in cancer. Consequently, altered WWOX levels have been observed in a wide variety of cancers. In vitro studies have identified a large number and variety of potential roles for WWOX. Although its normal role in vivo and functional contribution to cancer have not been fully defined, WWOX does have an integral role in metabolism and can suppress tumor growth. Using Drosophila melanogaster as an in vivo model system, we find that WWOX is a modulator of TNFα/Egr-mediated cell death. We found that altered levels of WWOX can modify phenotypes generated by low level ectopic expression of TNFα/Egr and this corresponds to altered levels of Caspase 3 activity. These results demonstrate an in vivo role for WWOX in promoting cell death. This form of cell death is accompanied by an increase in levels of reactive oxygen species, the regulation of which we have previously shown can also be modified by altered WWOX activity. We now hypothesise that, through regulation of reactive oxygen species, WWOX constitutes a link between alterations in cellular metabolism observed in cancer cells and their ability to evade normal cell death pathways. We have further shown that WWOX activity is required for the efficient removal of tumorigenic cells from a developing epithelial tissue. Together these results provide a molecular basis for the tumor suppressor functions of WWOX and the better prognosis observed in cancer patients with higher levels of WWOX activity. Understanding the conserved cellular pathways to which WWOX contributes provides novel possibilities for the development of therapeutic approaches to restore WWOX function in cancer.
Collapse
Affiliation(s)
- Louise V. O’Keefe
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Cheng Shoou Lee
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Amanda Choo
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Robert I. Richards
- Department of Genetics and Evolution, Centre for Molecular Pathology, School of Biological Sciences, The University of Adelaide, Adelaide, Australia
- * E-mail:
| |
Collapse
|
137
|
Cell mixing induced by myc is required for competitive tissue invasion and destruction. Nature 2015; 524:476-80. [DOI: 10.1038/nature14684] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/22/2015] [Indexed: 12/21/2022]
|
138
|
Wang Y, Antunes M, Anderson AE, Kadrmas JL, Jacinto A, Galko MJ. Integrin Adhesions Suppress Syncytium Formation in the Drosophila Larval Epidermis. Curr Biol 2015; 25:2215-27. [PMID: 26255846 DOI: 10.1016/j.cub.2015.07.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 06/15/2015] [Accepted: 07/10/2015] [Indexed: 12/16/2022]
Abstract
Integrins are critical for barrier epithelial architecture. Integrin loss in vertebrate skin leads to blistering and wound healing defects. However, how integrins and associated proteins maintain the regular morphology of epithelia is not well understood. We found that targeted knockdown of the integrin focal adhesion (FA) complex components β-integrin, PINCH, and integrin-linked kinase (ILK) caused formation of multinucleate epidermal cells within the Drosophila larval epidermis. This phenotype was specific to the integrin FA complex and not due to secondary effects on polarity or junctional structures. The multinucleate cells resembled the syncytia caused by physical wounding. Live imaging of wound-induced syncytium formation in the pupal epidermis suggested direct membrane breakdown leading to cell-cell fusion and consequent mixing of cytoplasmic contents. Activation of Jun N-terminal kinase (JNK) signaling, which occurs upon wounding, also correlated with syncytium formation induced by PINCH knockdown. Further, ectopic JNK activation directly caused epidermal syncytium formation. No mode of syncytium formation, including that induced by wounding, genetic loss of FA proteins, or local JNK hyperactivation, involved misregulation of mitosis or apoptosis. Finally, the mechanism of epidermal syncytium formation following JNK hyperactivation and wounding appeared to be direct disassembly of FA complexes. In conclusion, the loss-of-function phenotype of integrin FA components in the larval epidermis resembles a wound. Integrin FA loss in mouse and human skin also causes a wound-like appearance. Our results reveal a novel and unexpected role for proper integrin-based adhesion in suppressing larval epidermal cell-cell fusion--a role that may be conserved in other epithelia.
Collapse
Affiliation(s)
- Yan Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marco Antunes
- CEDOC-Faculdade de Ciências Médicas, Universidade Nova de Lisboa Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa Edificio Egas Moniz, Av Prof Egas Moniz, 1649-028 Lisboa, Portugal
| | - Aimee E Anderson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Julie L Kadrmas
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Antonio Jacinto
- CEDOC-Faculdade de Ciências Médicas, Universidade Nova de Lisboa Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; Instituto de Medicina Molecular Faculdade de Medicina da Universidade de Lisboa Edificio Egas Moniz, Av Prof Egas Moniz, 1649-028 Lisboa, Portugal; Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Michael J Galko
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Genes and Development Graduate Program, The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
139
|
Palanisamy R, Kumaresan V, Harikrishnan R, Arasu MV, Al-Dhabi NA, Arockiaraj J. Functional roles and gene regulation of tumor necrosis factor receptor 1 in freshwater striped murrel. Mol Immunol 2015; 66:240-252. [PMID: 25841174 DOI: 10.1016/j.molimm.2015.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/14/2015] [Accepted: 03/15/2015] [Indexed: 01/16/2023]
Abstract
In this study, a complete molecular characterization of tumor necrosis factor receptor 1 (TNFR1) which was identified from the constructed cDNA library of striped murrel Channa striatus (Cs) is reported. The CsTNFR1 encoded a type I membrane receptor protein that contains 399 amino acids including three cysteine-rich domains (CRDs) at CRD1(41-46), CRD2(79-118) and CRD3(120-159) in the extracellular region and a putative TNF receptor-associated factor (TRAF) site at 245-253 and a death domain between 297 and 388 in the cytoplasmic region which is essential for induction of apoptosis. The predicted molecular mass of CsTNFR1 is 45kDa and its corresponding theoretical isoelectric point (pI) is 6.3. CsTNFR1 shared maximum identity with TNFR1 from olive flounder Paralichthys olivaceus (82%). Real-time PCR showed that CsTNFR1 gene was expressed most abundantly (P<0.05) in the head kidney. Further, CsTNFR1 mRNA transcription was studied after challenge with fungus Apanomyces invadans and bacteria Aeromonas hydrophila. The fungus injected murrels showed a highest expression at 48h, whereas bacteria injected murrels showed at 24h. The gene expression studies revealed that CsTNFR1 may be involved in innate immune process of murrels against pathogenic infections. The over-expressed and purified recombinant CsTNFR1 protein (rCsTNFR1) was subjected to TNF-α inhibition assay to confirm their specificity and activity against TNF-α which confirmed that the rCsTNFR1 inhibits the activity of TNF-α in a dose dependent manner where maximum inhibition (97%) was observed at 10,000 fold concentration of rCsTNFR1. In addition, the direct cytotoxic effect of rCsTNFR1 was analyzed against head kidney phagocyte. The results showed that the recombinant CsTNFR1 protein does not exhibit any significant cytotoxicity against head kidney phagocyte cells even at higher concentration (8μg/ml). Moreover, the recombinant protein was analyzed for respiratory burst activity in the presence of two different ROS inducers, opsonized zymosan (fungal cell wall component) and phorbol 12-myristate 13-acetate (PMA). The findings showed that the C. striatus head kidney phagocyte exposed to purified recombinant CsTNFR1 protein alone do not produced any ROS. However, opsonized zymosan induced recombinant CsTNFR1 protein significantly (P<0.05) enhanced the ROS production on concentration basis which is revealed that the ROS production depends on the concentration of the recombinant CsTNFR1 protein. Overall, the study showed that the CsTNFR1 plays an important role in the pathogen-induced inflammatory process of striped murrel.
Collapse
MESH Headings
- Aeromonas hydrophila/immunology
- Amino Acid Sequence
- Animals
- Base Sequence
- Fish Diseases/immunology
- Fish Diseases/microbiology
- Fish Proteins/chemistry
- Fish Proteins/genetics
- Fish Proteins/immunology
- Fish Proteins/pharmacology
- Fishes/immunology
- Fishes/microbiology
- Gene Expression Regulation
- Gram-Negative Bacterial Infections/immunology
- Gram-Negative Bacterial Infections/microbiology
- Gram-Negative Bacterial Infections/veterinary
- Immunity, Innate
- Kidney/drug effects
- Kidney/immunology
- Kidney/microbiology
- Molecular Sequence Data
- Molecular Weight
- Mycoses/immunology
- Mycoses/microbiology
- Mycoses/veterinary
- Open Reading Frames
- Phagocytes/cytology
- Phagocytes/drug effects
- Phagocytes/immunology
- Protein Structure, Tertiary
- Reactive Oxygen Species/immunology
- Reactive Oxygen Species/metabolism
- Receptors, Tumor Necrosis Factor, Type I/chemistry
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Receptors, Tumor Necrosis Factor, Type I/pharmacology
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/pharmacology
- Sequence Alignment
- Tetradecanoylphorbol Acetate/pharmacology
- Zymosan/pharmacology
Collapse
Affiliation(s)
- Rajesh Palanisamy
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur 603 203, Chennai, Tamil Nadu, India
| | - Venkatesh Kumaresan
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur 603 203, Chennai, Tamil Nadu, India
| | - Ramasamy Harikrishnan
- Department of Zoology, Pachaiyappa's College for Men, Kanchipuram 631 501, Tamil Nadu, India
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Naif Abdullah Al-Dhabi
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Jesu Arockiaraj
- Division of Fisheries Biotechnology & Molecular Biology, Department of Biotechnology, Faculty of Science and Humanities, SRM University, Kattankulathur 603 203, Chennai, Tamil Nadu, India.
| |
Collapse
|
140
|
Enomoto M, Kizawa D, Ohsawa S, Igaki T. JNK signaling is converted from anti- to pro-tumor pathway by Ras-mediated switch of Warts activity. Dev Biol 2015; 403:162-71. [DOI: 10.1016/j.ydbio.2015.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 04/30/2015] [Accepted: 05/02/2015] [Indexed: 02/07/2023]
|
141
|
Tousled-like kinase mediated a new type of cell death pathway in Drosophila. Cell Death Differ 2015; 23:146-57. [PMID: 26088162 DOI: 10.1038/cdd.2015.77] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 04/21/2015] [Accepted: 05/05/2015] [Indexed: 01/09/2023] Open
Abstract
Programmed cell death (PCD) has an important role in sculpting organisms during development. However, much remains to be learned about the molecular mechanism of PCD. We found that ectopic expression of tousled-like kinase (tlk) in Drosophila initiated a new type of cell death. Furthermore, the TLK-induced cell death is likely to be independent of the canonical caspase pathway and other known caspase-independent pathways. Genetically, atg2 RNAi could rescue the TLK-induced cell death, and this function of atg2 was likely distinct from its role in autophagy. In the developing retina, loss of tlk resulted in reduced PCD in the interommatidial cells (IOCs). Similarly, an increased number of IOCs was present in the atg2 deletion mutant clones. However, double knockdown of tlk and atg2 by RNAi did not have a synergistic effect. These results suggested that ATG2 may function downstream of TLK. In addition to a role in development, tlk and atg2 RNAi could rescue calcium overload-induced cell death. Together, our results suggest that TLK mediates a new type of cell death pathway that occurs in both development and calcium cytotoxicity.
Collapse
|
142
|
Ma X, Xu W, Zhang D, Yang Y, Li W, Xue L. Wallenda regulates JNK-mediated cell death in Drosophila. Cell Death Dis 2015; 6:e1737. [PMID: 25950467 PMCID: PMC4669691 DOI: 10.1038/cddis.2015.111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/24/2015] [Accepted: 03/17/2015] [Indexed: 02/07/2023]
Abstract
The c-Jun N-terminal kinase (JNK) pathway plays essential roles in regulating a variety of cellular processes including proliferation, migration and survival. Previous genetic studies in Drosophila have identified numerous cell death regulating genes, providing new insights into the mechanisms for related diseases. Despite the known role of the small GTPase Rac1 in regulating cell death, the downstream components and underlying mechanism remain largely elusive. Here, we show that Rac1 promotes JNK-dependent cell death through Wallenda (Wnd). In addition, we find that Wnd triggers JNK activation and cell death via its kinase domain. Moreover, we show that both MKK4 and Hep are critical for Wnd-induced cell death. Furthermore, Wnd is essential for ectopic Egr- or Rho1-induced JNK activation and cell death. Finally, Wnd is physiologically required for loss of scribble-induced JNK-dependent cell death. Thus, our data suggest that wnd encodes a novel essential cell death regulator in Drosophila.
Collapse
Affiliation(s)
- X Ma
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - W Xu
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - D Zhang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Y Yang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - W Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - L Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, Shanghai 200092, China
| |
Collapse
|
143
|
Andersen DS, Colombani J, Palmerini V, Chakrabandhu K, Boone E, Röthlisberger M, Toggweiler J, Basler K, Mapelli M, Hueber AO, Léopold P. The Drosophila TNF receptor Grindelwald couples loss of cell polarity and neoplastic growth. Nature 2015; 522:482-6. [PMID: 25874673 DOI: 10.1038/nature14298] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/09/2015] [Indexed: 12/16/2022]
Abstract
Disruption of epithelial polarity is a key event in the acquisition of neoplastic growth. JNK signalling is known to play an important part in driving the malignant progression of many epithelial tumours, although the link between loss of polarity and JNK signalling remains elusive. In a Drosophila genome-wide genetic screen designed to identify molecules implicated in neoplastic growth, we identified grindelwald (grnd), a gene encoding a transmembrane protein with homology to members of the tumour necrosis factor receptor (TNFR) superfamily. Here we show that Grnd mediates the pro-apoptotic functions of Eiger (Egr), the unique Drosophila TNF, and that overexpression of an active form of Grnd lacking the extracellular domain is sufficient to activate JNK signalling in vivo. Grnd also promotes the invasiveness of Ras(V12)/scrib(-/-) tumours through Egr-dependent Matrix metalloprotease-1 (Mmp1) expression. Grnd localizes to the subapical membrane domain with the cell polarity determinant Crumbs (Crb) and couples Crb-induced loss of polarity with JNK activation and neoplastic growth through physical interaction with Veli (also known as Lin-7). Therefore, Grnd represents the first example of a TNFR that integrates signals from both Egr and apical polarity determinants to induce JNK-dependent cell death or tumour growth.
Collapse
Affiliation(s)
- Ditte S Andersen
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Genetics and Physiology of Growth laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Julien Colombani
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Genetics and Physiology of Growth laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Valentina Palmerini
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Krittalak Chakrabandhu
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Death receptors Signalling and Cancer Therapy laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Emilie Boone
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Genetics and Physiology of Growth laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Michael Röthlisberger
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Janine Toggweiler
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Konrad Basler
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Anne-Odile Hueber
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Death receptors Signalling and Cancer Therapy laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Pierre Léopold
- 1] University of Nice-Sophia Antipolis, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [2] CNRS, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [3] INSERM, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France [4] Genetics and Physiology of Growth laboratory, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| |
Collapse
|
144
|
Zhang S, Chen C, Wu C, Yang Y, Li W, Xue L. The canonical Wg signaling modulates Bsk-mediated cell death in Drosophila. Cell Death Dis 2015; 6:e1713. [PMID: 25855961 PMCID: PMC4650552 DOI: 10.1038/cddis.2015.85] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 12/21/2022]
Abstract
Cell death is an essential regulatory mechanism for removing unneeded cells in animal development and tissue homeostasis. The c-Jun N-terminal kinase (JNK) pathway has pivotal roles in the regulation of cell death in response to various intrinsic and extrinsic stress signals. The canonical Wingless (Wg) signaling has been implicated in cell proliferation and cell fate decisions, whereas its role in cell death remains largely elusive. Here, we report that activated Bsk (the Drosophila JNK homolog) induced cell death is mediated by the canonical Wg signaling. First, loss of Wg signaling abrogates Bsk-mediated caspase-independent cell death. Second, activation of Wg signaling promotes cell death in a caspase-independent manner. Third, activation of Bsk signaling results in upregulated transcription of wingless (wg) gene. Finally, Wg pathway participates in the physiological function of Bsk signaling in development. These findings not only reveal a previously undiscovered role of Wg signaling in Bsk-mediated cell death, but also provide a novel mechanism for the interplay between the two important signaling pathways in development.
Collapse
Affiliation(s)
- S Zhang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - C Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - C Wu
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Y Yang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - W Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - L Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| |
Collapse
|
145
|
Chittaranjan S, Xu J, Kuzyk M, Dullat HK, Wilton J, DeVorkin L, Lebovitz C, Morin GB, Marra MA, Gorski SM. The Drosophila TIPE family member Sigmar interacts with the Ste20-like kinase Misshapen and modulates JNK signaling, cytoskeletal remodeling and autophagy. Biol Open 2015; 4:672-84. [PMID: 25836674 PMCID: PMC4434819 DOI: 10.1242/bio.20148417] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
TNFAIP8 and other mammalian TIPE family proteins have attracted increased interest due to their associations with disease-related processes including oncogenic transformation, metastasis, and inflammation. The molecular and cellular functions of TIPE family proteins are still not well understood. Here we report the molecular and genetic characterization of the Drosophila TNFAIP8 homolog, CG4091/sigmar. Previous gene expression studies revealed dynamic expression of sigmar in larval salivary glands prior to histolysis. Here we demonstrate that in sigmar loss-of-function mutants, the salivary glands are morphologically abnormal with defects in the tubulin network and decreased autophagic flux. Sigmar localizes subcellularly to microtubule-containing projections in Drosophila S2 cells, and co-immunoprecipitates with the Ste20-like kinase Misshapen, a regulator of the JNK pathway. Further, the Drosophila TNF ligand Eiger can induce sigmar expression, and sigmar loss-of-function leads to altered localization of pDJNK in salivary glands. Together, these findings link Sigmar to the JNK pathway, cytoskeletal remodeling and autophagy activity during salivary gland development, and provide new insights into TIPE family member function.
Collapse
Affiliation(s)
- Suganthi Chittaranjan
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Jing Xu
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Michael Kuzyk
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Harpreet K Dullat
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada
| | - James Wilton
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Lindsay DeVorkin
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Chandra Lebovitz
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Gregg B Morin
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada Department of Medical Genetics, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Marco A Marra
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada
| | - Sharon M Gorski
- The Genome Sciences Centre, BC Cancer Agency, 675 West 10 Avenue, Vancouver, BC V5Z 1L3, Canada Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
146
|
Arias C, Fussero G, Zacharonok M, Macías A. Dpp-expressing and non-expressing cells: two different populations of growing cells in Drosophila. PLoS One 2015; 10:e0121457. [PMID: 25798905 PMCID: PMC4370563 DOI: 10.1371/journal.pone.0121457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/16/2015] [Indexed: 01/07/2023] Open
Abstract
There are different models that explain growth during development. One model is based on insect and amphibian regeneration studies. This model proposes that growth is directed by pattern, and growth takes place by intercalation at a growth discontinuity; therefore, proliferation should surround the discontinuity. Currently, this model, apart from regenerative studies on mostly adult patterning, has not found supporting evidence in Drosophila that shows proliferation surrounding a discontinuity. Despite this lack of evidence, the importance of discontinuities has been shown in different experiments, even under wt conditions, more specifically in the formation of the leg joints because of the occurrence of cell death at their boundaries. Here, we show the existence of a sharp discontinuity in Decapentaplegic (Dpp) in the genital discs at the third larvae stage (L3), which determines the upregulation in the Jun-NH2-Terminal-Kinase (JNK) pathway, reaper (rpr), head involution defective (hid) and active caspases from its boundaries. The proliferation and cell death surrounding the discontinuity suggest that growth can proceed by intercalation and competitive death takes place in this area. Finally, we show that the Rpr, Grim and Hid (RGH) products are a few of the factors that define the growth discontinuity because they are negative regulators of growth, a new function that is unique from their known functions in apoptosis.
Collapse
Affiliation(s)
- Carolina Arias
- Departamento de Fisiología, Cátedra de “Genética” Facultad de Ciencias Exactas Físicas y Naturales de la Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gimena Fussero
- Departamento de Fisiología, Cátedra de “Genética” Facultad de Ciencias Exactas Físicas y Naturales de la Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Marcelo Zacharonok
- Departamento de Fisiología, Cátedra de “Genética” Facultad de Ciencias Exactas Físicas y Naturales de la Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ana Macías
- Departamento de Fisiología, Cátedra de “Genética” Facultad de Ciencias Exactas Físicas y Naturales de la Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
147
|
Shklover J, Levy-Adam F, Kurant E. The role of Drosophila TNF Eiger in developmental and damage-induced neuronal apoptosis. FEBS Lett 2015; 589:871-9. [PMID: 25754009 DOI: 10.1016/j.febslet.2015.02.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/17/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022]
Abstract
Eiger, the sole Drosophila TNF-alpha homolog, causes ectopic apoptosis through JNK pathway activation. Yet, its role in developmental apoptosis remains unclear. eiger mutant flies are viable and fertile but display compromised elimination of oncogenic cells and extracellular bacteria. Here we show that Eiger, specifically expressed in embryonic neurons and glia, is not involved in developmental neuronal apoptosis or in apoptotic cell clearance. Instead, we provide evidence that Eiger is required for damage-induced apoptosis in the embryonic CNS through regulation of the pro-apoptotic gene hid independently of the JNK pathway. Our study thus reveals a new requirement for Eiger in eliminating damaged cells during development.
Collapse
Affiliation(s)
- Jeny Shklover
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Flonia Levy-Adam
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Estee Kurant
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
148
|
Shklover J, Mishnaevski K, Levy-Adam F, Kurant E. JNK pathway activation is able to synchronize neuronal death and glial phagocytosis in Drosophila. Cell Death Dis 2015; 6:e1649. [PMID: 25695602 PMCID: PMC4669801 DOI: 10.1038/cddis.2015.27] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 02/07/2023]
Abstract
Glial phagocytosis of superfluous neurons and damaged or aberrant neuronal material is crucial for normal development and maintenance of the CNS. However, the molecular mechanisms underlying the relationship between neuronal death and glial phagocytosis are poorly understood. We describe a novel mechanism that is able to synchronize neuronal cell death and glial phagocytosis of dying neurons in the Drosophila embryonic CNS. This mechanism involves c-Jun N-terminal kinase (JNK) signaling, which is required for developmental apoptosis of specific neurons during embryogenesis. We demonstrate that the dJNK pathway gain-of-function in neurons leads to dJNK signaling in glia, which results in upregulation of glial phagocytosis. Importantly, this promotion of phagocytosis is not mediated by upregulation of the glial phagocytic receptors SIMU and DRPR, but by increasing glial capacity to degrade apoptotic particles inside phagosomes. The proposed mechanism may be important for removal of damaged neurons in the developing and mature CNS.
Collapse
Affiliation(s)
- J Shklover
- Department of Genetics and Developmental Biology, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - K Mishnaevski
- Department of Genetics and Developmental Biology, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - F Levy-Adam
- Department of Genetics and Developmental Biology, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - E Kurant
- Department of Genetics and Developmental Biology, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
149
|
Chabu C, Xu T. Oncogenic Ras stimulates Eiger/TNF exocytosis to promote growth. Development 2014; 141:4729-39. [PMID: 25411211 DOI: 10.1242/dev.108092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oncogenic mutations in Ras deregulate cell death and proliferation to cause cancer in a significant number of patients. Although normal Ras signaling during development has been well elucidated in multiple organisms, it is less clear how oncogenic Ras exerts its effects. Furthermore, cancers with oncogenic Ras mutations are aggressive and generally resistant to targeted therapies or chemotherapy. We identified the exocytosis component Sec15 as a synthetic suppressor of oncogenic Ras in an in vivo Drosophila mosaic screen. We found that oncogenic Ras elevates exocytosis and promotes the export of the pro-apoptotic ligand Eiger (Drosophila TNF). This blocks tumor cell death and stimulates overgrowth by activating the JNK-JAK-STAT non-autonomous proliferation signal from the neighboring wild-type cells. Inhibition of Eiger/TNF exocytosis or interfering with the JNK-JAK-STAT non-autonomous proliferation signaling at various steps suppresses oncogenic Ras-mediated overgrowth. Our findings highlight important cell-intrinsic and cell-extrinsic roles of exocytosis during oncogenic growth and provide a new class of synthetic suppressors for targeted therapy approaches.
Collapse
Affiliation(s)
- Chiswili Chabu
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, Boyer Center for Molecular Medicine, 295 Congress Avenue, New Haven, CT 06536, USA
| | - Tian Xu
- Department of Genetics, Howard Hughes Medical Institute, Yale University School of Medicine, Boyer Center for Molecular Medicine, 295 Congress Avenue, New Haven, CT 06536, USA
| |
Collapse
|
150
|
Takino K, Ohsawa S, Igaki T. Loss of Rab5 drives non-autonomous cell proliferation through TNF and Ras signaling in Drosophila. Dev Biol 2014; 395:19-28. [PMID: 25224221 DOI: 10.1016/j.ydbio.2014.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/26/2014] [Accepted: 09/05/2014] [Indexed: 01/16/2023]
Abstract
Deregulation of the endocytic machinery has been implicated in human cancers. However, the mechanism by which endocytic defects drive cancer development remains to be clarified. Here, we find through a genetic screen in Drosophila that loss of Rab5, a protein required for early endocytic trafficking, drives non-autonomous cell proliferation in imaginal epithelium. Our genetic data indicate that dysfunction of Rab5 leads to cell-autonomous accumulation of Eiger (a TNF homolog) and EGF receptor (EGFR), which causes activation of downstream JNK and Ras signaling, respectively. JNK signaling and its downstream component Cdc42 cooperate with Ras signaling to induce upregulation of a secreted growth factor Upd (an IL-6 homolog) through inactivation of the Hippo pathway. Such non-autonomous tissue growth triggered by Rab5 defect could contribute to epithelial homeostasis as well as cancer development within heterogeneous tumor microenvironment.
Collapse
Affiliation(s)
- Kyoko Takino
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; Division of Genetics, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Shizue Ohsawa
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho-cho, Sakyo-ku, Kyoto 606-8501, Japan; PRESTO, Japan Science and Technology Agency (JST), 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|