51
|
Pinto-Teixeira F, Konstantinides N, Desplan C. Programmed cell death acts at different stages of Drosophila neurodevelopment to shape the central nervous system. FEBS Lett 2016; 590:2435-2453. [PMID: 27404003 DOI: 10.1002/1873-3468.12298] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 12/19/2022]
Abstract
Nervous system development is a process that integrates cell proliferation, differentiation, and programmed cell death (PCD). PCD is an evolutionary conserved mechanism and a fundamental developmental process by which the final cell number in a nervous system is established. In vertebrates and invertebrates, PCD can be determined intrinsically by cell lineage and age, as well as extrinsically by nutritional, metabolic, and hormonal states. Drosophila has been an instrumental model for understanding how this mechanism is regulated. We review the role of PCD in Drosophila central nervous system development from neural progenitors to neurons, its molecular mechanism and function, how it is regulated and implemented, and how it ultimately shapes the fly central nervous system from the embryo to the adult. Finally, we discuss ideas that emerged while integrating this information.
Collapse
Affiliation(s)
- Filipe Pinto-Teixeira
- Department of Biology, New York University 1009 Silver Center 100 Washington Square East, New York, NY 10003, USA.,Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi 129188, UAE
| | - Nikolaos Konstantinides
- Department of Biology, New York University 1009 Silver Center 100 Washington Square East, New York, NY 10003, USA
| | - Claude Desplan
- Department of Biology, New York University 1009 Silver Center 100 Washington Square East, New York, NY 10003, USA.,Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi 129188, UAE
| |
Collapse
|
52
|
Farkaš R, Pečeňová L, Mentelová L, Beňo M, Beňová-Liszeková D, Mahmoodová S, Tejnecký V, Raška O, Juda P, Svidenská S, Hornáček M, Chase BA, Raška I. Massive excretion of calcium oxalate from late prepupal salivary glands of Drosophila melanogaster demonstrates active nephridial-like anion transport. Dev Growth Differ 2016; 58:562-74. [PMID: 27397870 DOI: 10.1111/dgd.12300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 04/24/2016] [Accepted: 05/16/2016] [Indexed: 02/01/2023]
Abstract
The Drosophila salivary glands (SGs) were well known for the puffing patterns of their polytene chromosomes and so became a tissue of choice to study sequential gene activation by the steroid hormone ecdysone. One well-documented function of these glands is to produce a secretory glue, which is released during pupariation to fix the freshly formed puparia to the substrate. Over the past two decades SGs have been used to address specific aspects of developmentally-regulated programmed cell death (PCD) as it was thought that they are doomed for histolysis and after pupariation are just awaiting their fate. More recently, however, we have shown that for the first 3-4 h after pupariation SGs undergo tremendous endocytosis and vacuolation followed by vacuole neutralization and membrane consolidation. Furthermore, from 8 to 10 h after puparium formation (APF) SGs display massive apocrine secretion of a diverse set of cellular proteins. Here, we show that during the period from 11 to 12 h APF, the prepupal glands are very active in calcium oxalate (CaOx) extrusion that resembles renal or nephridial excretory activity. We provide genetic evidence that Prestin, a Drosophila homologue of the mammalian electrogenic anion exchange carrier SLC26A5, is responsible for the instantaneous production of CaOx by the late prepupal SGs. Its positive regulation by the protein kinases encoded by fray and wnk lead to increased production of CaOx. The formation of CaOx appears to be dependent on the cooperation between Prestin and the vATPase complex as treatment with bafilomycin A1 or concanamycin A abolishes the production of detectable CaOx. These data demonstrate that prepupal SGs remain fully viable, physiologically active and engaged in various cellular activities at least until early pupal period, that is, until moments prior to the execution of PCD.
Collapse
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Ludmila Pečeňová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia.,Department of Genetics, Comenius University, Mlynská dolina B-1, 84215, Bratislava, Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia.,Department of Genetics, Comenius University, Mlynská dolina B-1, 84215, Bratislava, Slovakia
| | - Milan Beňo
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
| | - Silvia Mahmoodová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Centre, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia.,Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Malá Hora 4, 03601, Martin, Slovakia
| | - Václav Tejnecký
- Faculty of Agrobiology, Food and Natural Resources, Czech Agricultural University, Kamýcká 129, 16521, Prague 6, Czech Republic
| | - Otakar Raška
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Albertov 4, 12800, Prague, Czech Republic
| | - Pavel Juda
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Albertov 4, 12800, Prague, Czech Republic
| | - Silvie Svidenská
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Albertov 4, 12800, Prague, Czech Republic
| | - Matúš Hornáček
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Albertov 4, 12800, Prague, Czech Republic
| | - Bruce A Chase
- Department of Biology, University of Nebraska at Omaha, 6001 Dodge Street, Omaha, Nebraska, 68182-0040, USA
| | - Ivan Raška
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Albertov 4, 12800, Prague, Czech Republic
| |
Collapse
|
53
|
Ding X, Sun G, Argaw YG, Wong JO, Easwaran S, Montell DJ. CasExpress reveals widespread and diverse patterns of cell survival of caspase-3 activation during development in vivo. eLife 2016; 5:e10936. [PMID: 27058168 PMCID: PMC4865370 DOI: 10.7554/elife.10936] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 04/08/2016] [Indexed: 02/06/2023] Open
Abstract
Caspase-3 carries out the executioner phase of apoptosis, however under special circumstances, cells can survive its activity. To document systematically where and when cells survive caspase-3 activation in vivo, we designed a system, CasExpress, which drives fluorescent protein expression, transiently or permanently, in cells that survive caspase-3 activation in Drosophila. We discovered widespread survival of caspase-3 activity. Distinct spatial and temporal patterns emerged in different tissues. Some cells activated caspase-3 during their normal development in every cell and in every animal without evidence of apoptosis. In other tissues, such as the brain, expression was sporadic both temporally and spatially and overlapped with periods of apoptosis. In adults, reporter expression was evident in a large fraction of cells in most tissues of every animal; however the precise patterns varied. Inhibition of caspase activity in wing discs reduced wing size demonstrating functional significance. The implications of these patterns are discussed.
Collapse
Affiliation(s)
- Xun Ding
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
- Department of Biological Chemistry, Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, United States
| | - Gongping Sun
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
| | - Yewubdar G Argaw
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
| | - Jessica O Wong
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
| | - Sreesankar Easwaran
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
| | - Denise J Montell
- Molecular, Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, United States
- Department of Biological Chemistry, Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, United States
| |
Collapse
|
54
|
Ehrhardt E, Graf P, Kleele T, Liu Y, Boyan G. Fates of identified pioneer cells in the developing antennal nervous system of the grasshopper Schistocerca gregaria. ARTHROPOD STRUCTURE & DEVELOPMENT 2016; 45:23-30. [PMID: 26597904 DOI: 10.1016/j.asd.2015.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/06/2015] [Accepted: 11/06/2015] [Indexed: 06/05/2023]
Abstract
In the early embryonic grasshopper, two pairs of sibling cells near the apex of the antenna pioneer its dorsal and ventral nerve tracts to the brain. En route, the growth cones of these pioneers contact a so-called base pioneer associated with each tract and which acts as a guidepost cell. Both apical and basal pioneers express stereotypic molecular labels allowing them to be uniquely identified. Although their developmental origins are largely understood, the fates of the respective pioneers remain unclear. We therefore employed the established cell death markers acridine orange and TUNEL to determine whether the apical and basal pioneers undergo apoptosis during embryogenesis. Our data reveal that the apical pioneers maintain a consistent molecular profile from their birth up to mid-embryogenesis, at which point the initial antennal nerve tracts to the brain have been established. Shortly after this the apical pioneers undergo apoptosis. Death occurs at a developmental stage similar to that reported elsewhere for pioneers in a leg - an homologous appendage. Base pioneers, by contrast, progressively change their molecular profile and can no longer be unequivocally identified after mid-embryogenesis. At no stage up to then do they exhibit death labels. If they persist, the base pioneers must be assumed to adopt a new role in the developing antennal nervous system.
Collapse
Affiliation(s)
- Erica Ehrhardt
- Graduate School of Systemic Neuroscience, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany
| | - Philip Graf
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany
| | - Tatjana Kleele
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany; Institute of Neuronal Cell Biology, Technische Universität München, Biedersteinerstr. 29, 80801, Munich, Germany
| | - Yu Liu
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany
| | - George Boyan
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany.
| |
Collapse
|
55
|
|
56
|
Li Y, Wang X, Hou Y, Zhou X, Chen Q, Guo C, Xia Q, Zhang Y, Zhao P. Integrative Proteomics and Metabolomics Analysis of Insect Larva Brain: Novel Insights into the Molecular Mechanism of Insect Wandering Behavior. J Proteome Res 2015; 15:193-204. [PMID: 26644297 DOI: 10.1021/acs.jproteome.5b00736] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Before metamorphosis, most holometabolous insects, such as the silkworm studied here, undergo a special phase called the wandering stage. Insects in this stage often display enhanced locomotor activity (ELA). ELA is vital because it ensures that the insect finds a safe and suitable place to live through the pupal stage. The physiological mechanisms of wandering behavior are still unclear. Here, we integrated proteomics and metabolomics approaches to analyze the brain of the lepidopteran insect, silkworm, at the feeding and wandering stages. Using LC-MS/MS and GC-MS, in all we identified 3004 proteins and 37 metabolites at these two stages. Among them, 465 proteins and 22 metabolites were changed. Neural signal transduction proteins and metabolites, such as neurofilament, dopaminergic synapse related proteins, and glutamic acid, were significantly altered, which suggested that active neural conduction occurred in the brain at the wandering stage. We also found decreased dopamine degradation at the wandering stage. The proposed changes in active neural conduction and increased dopamine concentration might induce ELA. In addition, proteins involved in the ubiquitin proteasome system and lysosome pathway were upregulated, revealing that the brain experiences morphological remodeling during metamorphosis. These findings yielded novel insights into the molecular mechanism underlying insect wandering behavior.
Collapse
Affiliation(s)
- Yi Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Xin Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Yong Hou
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Xiaoying Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Quanmei Chen
- Department of Biochemistry & Molecular Biology, Chongqing Medical University , Chongqing 400016, China
| | - Chao Guo
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Yan Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| |
Collapse
|
57
|
Clavier A, Rincheval-Arnold A, Colin J, Mignotte B, Guénal I. Apoptosis in Drosophila: which role for mitochondria? Apoptosis 2015; 21:239-51. [DOI: 10.1007/s10495-015-1209-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
58
|
Goto T, Sato K, Sone H, Koganezawa M, Ito H, Yamamoto D. Zeste tunes the timing of ecdysone actions in triggering programmed tissue degeneration in Drosophila. J Neurogenet 2015; 29:169-73. [PMID: 26577029 DOI: 10.3109/01677063.2015.1098638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In the pupal stage, the fly body undergoes extensive metamorphic remodeling, in which programmed cell death plays a critical role. We studied two of the constituent processes in this remodeling, salivary gland degeneration and breakdown of the eclosion muscle, which are triggered by an increase and a decrease in the circulating steroid hormone ecdysone at the start and end of metamorphosis, respectively. We found that knockdown of zeste (z), a gene encoding a sequence-specific DNA-binding protein implicated in transvection, in salivary gland cells advances the initiation of their degeneration, whereas z knockdown in neurons delays muscle breakdown. We further showed that knockdown of an ecdysone-inducible gene, E74, retards salivary gland degeneration with little effect on eclosion muscle breakdown. We propose that Z tunes the sensitivity of ecdysone targets to this hormone in order to ensure a high safety margin so that the cell death program will be activated when the ecdysone titer is at a sufficiently high level that is reached only at a defined stage during metamorphosis.
Collapse
Affiliation(s)
- Takaaki Goto
- a Division of Neurogenetics , Tohoku University Graduate School of Life Sciences , Sendai , Japan and
| | - Kosei Sato
- a Division of Neurogenetics , Tohoku University Graduate School of Life Sciences , Sendai , Japan and
| | - Hiroyuki Sone
- b Laboratory of Genetics , Waseda University School of Human Sciences, Nishi-Tokyo , Tokyo , Japan
| | - Masayuki Koganezawa
- a Division of Neurogenetics , Tohoku University Graduate School of Life Sciences , Sendai , Japan and
| | - Hiroki Ito
- a Division of Neurogenetics , Tohoku University Graduate School of Life Sciences , Sendai , Japan and.,b Laboratory of Genetics , Waseda University School of Human Sciences, Nishi-Tokyo , Tokyo , Japan
| | - Daisuke Yamamoto
- a Division of Neurogenetics , Tohoku University Graduate School of Life Sciences , Sendai , Japan and.,b Laboratory of Genetics , Waseda University School of Human Sciences, Nishi-Tokyo , Tokyo , Japan
| |
Collapse
|
59
|
Urbanek A, Richert M, Kapusta M. Metamorphic changes in abdominal spines of Forcipomyia nigra pupae (Diptera: Ceratopogonidae). ARTHROPOD STRUCTURE & DEVELOPMENT 2015; 44:554-567. [PMID: 26297424 DOI: 10.1016/j.asd.2015.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 06/04/2023]
Abstract
Pupae of Forcipomyia nigra biting midges bear double rows of dorsal and lateral spines. Their arrangement corresponds to the distribution of larval mechanosensory setae. They are serrated simple cuticular structures with tubercles but, in contrast to larval secretory mechanoreceptors, they are not innervated and do not exhibit any pores. The ultrastructure of abdominal spines varies among different pupal stages. They are produced by epidermal cells which fill the interior of the spine. In the youngest pupae epidermal cells are tightly packed and adhere to the cuticle. Then, the cells withdraw from the spinal cavity and the beginning of autophagy is observed. The last stage represents abdominal spines without any cellular material and then apoptosis probably proceeds in the withdrawn epidermal cells. Since the pupal spines occupied the same region of the segment as the larval setae, we consider that the same genes are responsible for their formation as for the formation of epidermal cells but that their mechanosensory and secretory function is no longer needed.
Collapse
Affiliation(s)
- Aleksandra Urbanek
- Department of Invertebrate Zoology and Parasitology, University of Gdańsk, ul. Wita Stwosza 59, 80-308 Gdańsk, Poland.
| | - Malwina Richert
- Laboratory of Electron Microscopy, University of Gdańsk, ul. Wita Stwosza 59, 80-308 Gdańsk, Poland
| | - Małgorzata Kapusta
- Department of Plant Cytology and Embryology, University of Gdańsk, ul. Wita Stwosza 59, 80-308 Gdańsk, Poland
| |
Collapse
|
60
|
Farkaš R, Sláma K. Respiratory metabolism of salivary glands during the late larval and prepupal development of Drosophila melanogaster. JOURNAL OF INSECT PHYSIOLOGY 2015; 81:109-117. [PMID: 26116777 DOI: 10.1016/j.jinsphys.2015.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 06/04/2023]
Abstract
During the late larval period, the salivary glands (SG) of Drosophila show a cascade of cytological changes associated with exocytosis and the expectoration of the proteinaceous glue that is used to affix the pupariating larva to a substrate. After puparium formation (APF), SG undergo extensive cytoplasmic vacuolation due to endocytosis, vacuole consolidation and massive apocrine secretion. Here we investigated possible correlations between cytological changes, the puffing pattern in polytene chromosomes and respiratory metabolism of the SG. The carefully staged SG were explanted into small amounts (1 or 2μl) of tissue culture medium. The respiratory metabolism of single or up to 3 pairs of glands was evaluated by recording the rate of O2 consumption using a scanning microrespirographic technique sensitive to subnanoliter volumes of the respiratory O2 or CO2. The recordings were carried out at times between 8h before pupariation (BPF), until 16h APF, at which point the SG completely disintegrate. At the early wandering larval stage (8h BPF), the glands consume 2nl of O2/gland/min (=2500μl O2/g/h). This relatively high metabolic rate decreases down to 1.2-1.3nl of O2 during the endogenous peak in ecdysteroid concentration that culminates around pupariation. The metabolic decline coincides with the exocytosis of the proteinaceous glue. During and shortly after puparium formation, which is accompanied cytologically by intense vacuolation, O2 consumption in the SG temporarily increases to 1.6nl O2/gland/min. After this time, the metabolic rate of the SG decreases downward steadily until 16h APF, when the glands disintegrate and cease to consume oxygen. The SG we analyzed from Drosophila larvae were composed of 134 intrinsic cells, with the average volume of one lobe being 37nl. Therefore, a single SG cell of the wandering larva (with O2 consumption of 2nl/gland/min), consumes each about 16pl of O2/cell/min. A simultaneous analysis of the rate of protein and RNA synthesis in the SG shows a course similar to that found in respiratory metabolism.
Collapse
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlárska 3, 833 06 Bratislava, Slovakia.
| | - Karel Sláma
- Institute of Entomology, Czech Academy of Sciences, Drnovská 507, 161 00 Prague 6 - Ruzyně, Czech Republic
| |
Collapse
|
61
|
Pro-apoptotic gene regulation and its activation by gamma-irradiation in the Caribbean fruit fly, Anastrepha suspensa. Apoptosis 2015; 20:1-9. [PMID: 25433919 DOI: 10.1007/s10495-014-1055-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Transcriptional activation of pro-apoptotic genes in response to cytotoxic stimuli is a conserved feature of the cell death pathway in metazoans. However, understanding the extent of this conservation in insects has been limited by the lack of known pro-apoptotic genes in non-drosophilids. Recently, we described the pro-apoptotic genes, Asrpr and Ashid, from the tephritid, Anastrepha suspensa, that now allow us to explore the conservation of pro-apoptotic gene regulation between a tephritid and drosophilids. In this study, we determined the developmental profiles of Asrpr and Ashid transcripts during embryogenesis and in embryos exposed to γ-irradiation. Transcript levels of both genes determined by qRT-PCR were low throughout embryogenesis, with strong Ashid expression occurring during early to mid-embryogenesis and Asrpr expression peaking in late embryogenesis. This correlated to acridine orange stained apoptotic cells first appearing at 17 h and increasing over time. However, when irradiated at 16 h post-oviposition embryos exhibited significant levels of apoptosis consistent with strong induction of Asrpr and Ashid transcript levels by γ-irradiation in young embryos <24 h post-oviposition. Furthermore, embryos irradiated <24 h post-oviposition failed to hatch, those irradiated between 24 and 32 h had increased hatching rates, but between 48 and 72 h irradiation had no effect on egg hatching. This indicates a transition of embryos from an irradiation-sensitive to irradiation-resistance stage between 24 and 48 h. Throughout post-embryonic development, the two pro-apoptotic genes share similar patterns of up-regulated gene expression, which correlate to ecdysone-induced developmental events, especially during metamorphosis. Together these results provide the first direct evidence for a conserved molecular mechanism of the programmed cell death pathway in insects.
Collapse
|
62
|
Dong DJ, Jing YP, Liu W, Wang JX, Zhao XF. The Steroid Hormone 20-Hydroxyecdysone Up-regulates Ste-20 Family Serine/Threonine Kinase Hippo to Induce Programmed Cell Death. J Biol Chem 2015; 290:24738-46. [PMID: 26272745 DOI: 10.1074/jbc.m115.643783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Indexed: 12/15/2022] Open
Abstract
The steroid hormone 20-hydroxyecdysone (20E) and the serine/threonine Ste20-like kinase Hippo signal promote programmed cell death (PCD) during development, although the interaction between them remains unclear. Here, we present evidence that 20E up-regulates Hippo to induce PCD during the metamorphic development of insects. We found that Hippo is involved in 20E-induced metamorphosis via promoting the phosphorylation and cytoplasmic retention of Yorkie (Yki), causing suppressed expression of the inhibitor of apoptosis (IAP), thereby releasing its inhibitory effect on caspase. Furthermore, we show that 20E induced the expression of Hippo at the transcriptional level through the ecdysone receptor (EcR), ultraspiracle protein (USP), and hormone receptor 3 (HR3). We also found that Hippo suppresses the binding of Yki complex to the HR3 promoter. In summary, 20E up-regulates the transcription of Hippo via EcRB1, USP1, and HR3 to induce PCD, and Hippo has negative feedback effects on HR3 expression. These two signaling pathways coordinate PCD during insect metamorphosis.
Collapse
Affiliation(s)
- Du-Juan Dong
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Yu-Pu Jing
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Wen Liu
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Jin-Xing Wang
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Xiao-Fan Zhao
- From the Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan 250100, China
| |
Collapse
|
63
|
Novel Genes Involved in Controlling Specification of Drosophila FMRFamide Neuropeptide Cells. Genetics 2015; 200:1229-44. [PMID: 26092715 DOI: 10.1534/genetics.115.178483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/16/2015] [Indexed: 01/28/2023] Open
Abstract
The expression of neuropeptides is often extremely restricted in the nervous system, making them powerful markers for addressing cell specification . In the developing Drosophila ventral nerve cord, only six cells, the Ap4 neurons, of some 10,000 neurons, express the neuropeptide FMRFamide (FMRFa). Each Ap4/FMRFa neuron is the last-born cell generated by an identifiable and well-studied progenitor cell, neuroblast 5-6 (NB5-6T). The restricted expression of FMRFa and the wealth of information regarding its gene regulation and Ap4 neuron specification makes FMRFa a valuable readout for addressing many aspects of neural development, i.e., spatial and temporal patterning cues, cell cycle control, cell specification, axon transport, and retrograde signaling. To this end, we have conducted a forward genetic screen utilizing an Ap4-specific FMRFa-eGFP transgenic reporter as our readout. A total of 9781 EMS-mutated chromosomes were screened for perturbations in FMRFa-eGFP expression, and 611 mutants were identified. Seventy-nine of the strongest mutants were mapped down to the affected gene by deficiency mapping or whole-genome sequencing. We isolated novel alleles for previously known FMRFa regulators, confirming the validity of the screen. In addition, we identified novel essential genes, including several with previously undefined functions in neural development. Our identification of genes affecting most major steps required for successful terminal differentiation of Ap4 neurons provides a comprehensive view of the genetic flow controlling the generation of highly unique neuronal cell types in the developing nervous system.
Collapse
|
64
|
Live to die another way: modes of programmed cell death and the signals emanating from dying cells. Nat Rev Mol Cell Biol 2015; 16:329-44. [PMID: 25991373 DOI: 10.1038/nrm3999] [Citation(s) in RCA: 477] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
All life ends in death, but perhaps one of life's grander ironies is that it also depends on death. Cell-intrinsic suicide pathways, termed programmed cell death (PCD), are crucial for animal development, tissue homeostasis and pathogenesis. Originally, PCD was almost synonymous with apoptosis; recently, however, alternative mechanisms of PCD have been reported. Here, we provide an overview of several distinct PCD mechanisms, namely apoptosis, autophagy and necroptosis. In addition, we discuss the complex signals that emanate from dying cells, which can either trigger regeneration or instruct additional killing. Further advances in understanding the physiological roles of the various mechanisms of cell death and their associated signals will be important to selectively manipulate PCD for therapeutic purposes.
Collapse
|
65
|
Accorsi A, Zibaee A, Malagoli D. The multifaceted activity of insect caspases. JOURNAL OF INSECT PHYSIOLOGY 2015; 76:17-23. [PMID: 25783954 DOI: 10.1016/j.jinsphys.2015.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/09/2015] [Accepted: 03/09/2015] [Indexed: 06/04/2023]
Abstract
Caspases are frequently considered synonymous with apoptotic cell death. Increasing evidence demonstrates that these proteases may exert their activities in non-apoptotic functions. The non-apoptotic roles of caspases may include developmentally regulated autophagy during insect metamorphosis, as well as neuroblast self-renewal and the immune response. Here, we summarize the established knowledge and the recent advances in the multiple roles of insect caspases to highlight their relevance for physiological processes and survival.
Collapse
Affiliation(s)
- A Accorsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A Zibaee
- Department of Plant Protection, Faculty of Agricultural Sciences, University of Guilan, Rasht, Iran
| | - D Malagoli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
66
|
Tait SWG, Ichim G, Green DR. Die another way--non-apoptotic mechanisms of cell death. J Cell Sci 2015; 127:2135-44. [PMID: 24833670 DOI: 10.1242/jcs.093575] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Regulated, programmed cell death is crucial for all multicellular organisms. Cell death is essential in many processes, including tissue sculpting during embryogenesis, development of the immune system and destruction of damaged cells. The best-studied form of programmed cell death is apoptosis, a process that requires activation of caspase proteases. Recently it has been appreciated that various non-apoptotic forms of cell death also exist, such as necroptosis and pyroptosis. These non-apoptotic cell death modalities can be either triggered independently of apoptosis or are engaged should apoptosis fail to execute. In this Commentary, we discuss several regulated non-apoptotic forms of cell death including necroptosis, autophagic cell death, pyroptosis and caspase-independent cell death. We outline what we know about their mechanism, potential roles in vivo and define outstanding questions. Finally, we review data arguing that the means by which a cell dies actually matters, focusing our discussion on inflammatory aspects of cell death.
Collapse
Affiliation(s)
- Stephen W G Tait
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Gabriel Ichim
- Cancer Research UK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Switchback Road, Glasgow G61 1BD, UK
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
67
|
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
|
68
|
Sopko R, Lin YB, Makhijani K, Alexander B, Perrimon N, Brückner K. A systems-level interrogation identifies regulators of Drosophila blood cell number and survival. PLoS Genet 2015; 11:e1005056. [PMID: 25749252 PMCID: PMC4352040 DOI: 10.1371/journal.pgen.1005056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 02/05/2015] [Indexed: 12/12/2022] Open
Abstract
In multicellular organisms, cell number is typically determined by a balance of intracellular signals that positively and negatively regulate cell survival and proliferation. Dissecting these signaling networks facilitates the understanding of normal development and tumorigenesis. Here, we study signaling by the Drosophila PDGF/VEGF Receptor (Pvr) in embryonic blood cells (hemocytes) and in the related cell line Kc as a model for the requirement of PDGF/VEGF receptors in vertebrate cell survival and proliferation. The system allows the investigation of downstream and parallel signaling networks, based on the ability of Pvr to activate Ras/Erk, Akt/TOR, and yet-uncharacterized signaling pathway/s, which redundantly mediate cell survival and contribute to proliferation. Using Kc cells, we performed a genome wide RNAi screen for regulators of cell number in a sensitized, Pvr deficient background. We identified the receptor tyrosine kinase (RTK) Insulin-like receptor (InR) as a major Pvr Enhancer, and the nuclear hormone receptors Ecdysone receptor (EcR) and ultraspiracle (usp), corresponding to mammalian Retinoid X Receptor (RXR), as Pvr Suppressors. In vivo analysis in the Drosophila embryo revealed a previously unrecognized role for EcR to promote apoptotic death of embryonic blood cells, which is balanced with pro-survival signaling by Pvr and InR. Phosphoproteomic analysis demonstrates distinct modes of cell number regulation by EcR and RTK signaling. We define common phosphorylation targets of Pvr and InR that include regulators of cell survival, and unique targets responsible for specialized receptor functions. Interestingly, our analysis reveals that the selection of phosphorylation targets by signaling receptors shows qualitative changes depending on the signaling status of the cell, which may have wide-reaching implications for other cell regulatory systems.
Collapse
Affiliation(s)
- Richelle Sopko
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - You Bin Lin
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Kalpana Makhijani
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Brandy Alexander
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Boston, Massachusetts, United States of America
| | - Katja Brückner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
69
|
Eichner C, Dalvin S, Skern-Mauritzen R, Malde K, Kongshaug H, Nilsen F. Characterization of a novel RXR receptor in the salmon louse (Lepeophtheirus salmonis, Copepoda) regulating growth and female reproduction. BMC Genomics 2015; 16:81. [PMID: 25765704 PMCID: PMC4333900 DOI: 10.1186/s12864-015-1277-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/22/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nuclear receptors have crucial roles in all metazoan animals as regulators of gene transcription. A wide range of studies have elucidated molecular and biological significance of nuclear receptors but there are still a large number of animals where the knowledge is very limited. In the present study we have identified an RXR type of nuclear receptor in the salmon louse (Lepeophtheirus salmonis) (i.e. LsRXR). RXR is one of the two partners of the Ecdysteroid receptor in arthropods, the receptor for the main molting hormone 20-hydroxyecdysone (E20) with a wide array of effects in arthropods. RESULTS Five different LsRXR transcripts were identified by RACE showing large differences in domain structure. The largest isoforms contained complete DNA binding domain (DBD) and ligand binding domain (LBD), whereas some variants had incomplete or no DBD. LsRXR is transcribed in several tissues in the salmon louse including ovary, subcuticular tissue, intestine and glands. By using Q-PCR it is evident that the LsRXR mRNA levels vary throughout the L. salmonis life cycle. We also show that the truncated LsRXR transcript comprise about 50% in all examined samples. We used RNAi to knock-down the transcription in adult reproducing female lice. This resulted in close to zero viable offspring. We also assessed the LsRXR RNAi effects using a L. salmonis microarray and saw significant effects on transcription in the female lice. Transcription of the major yolk proteins was strongly reduced by knock-down of LsRXR. Genes involved in lipid metabolism and transport were also down regulated. Furthermore, different types of growth processes were up regulated and many cuticle proteins were present in this group. CONCLUSIONS The present study demonstrates the significance of LsRXR in adult female L. salmonis and discusses the functional aspects in relation to other arthropods. LsRXR has a unique structure that should be elucidated in the future.
Collapse
Affiliation(s)
- Christiane Eichner
- Department of Biology, Sea Lice Research Centre, University of Bergen, Bergen, Norway.
| | - Sussie Dalvin
- Department of Biology, Sea Lice Research Centre, University of Bergen, Bergen, Norway. .,Institute of Marine Research, Bergen, Norway.
| | | | - Ketil Malde
- Institute of Marine Research, Bergen, Norway.
| | - Heidi Kongshaug
- Department of Biology, Sea Lice Research Centre, University of Bergen, Bergen, Norway.
| | - Frank Nilsen
- Department of Biology, Sea Lice Research Centre, University of Bergen, Bergen, Norway.
| |
Collapse
|
70
|
Cell death in development: Signaling pathways and core mechanisms. Semin Cell Dev Biol 2015; 39:12-9. [PMID: 25668151 DOI: 10.1016/j.semcdb.2015.02.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/22/2015] [Accepted: 02/02/2015] [Indexed: 11/20/2022]
Abstract
Programmed cell death eliminates unneeded and dangerous cells in a timely and effective manner during development. In this review, we examine the role cell death plays during development in worms, flies and mammals. We discuss signaling pathways that regulate developmental cell death, and describe how they communicate with the core cell death pathways. In most organisms, the majority of developmental cell death is seen in the nervous system. Therefore we focus on what is known about the regulation of developmental cell death in this tissue. Understanding how the cell death is regulated during development may provide insight into how this process can be manipulated in the treatment of disease.
Collapse
|
71
|
Lin L, Baehrecke EH. Autophagy, cell death, and cancer. Mol Cell Oncol 2015; 2:e985913. [PMID: 27308466 PMCID: PMC4905302 DOI: 10.4161/23723556.2014.985913] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 12/11/2022]
Abstract
Autophagy is an evolutionarily conserved intracellular catabolic process that is used by all cells to degrade dysfunctional or unnecessary cytoplasmic components through delivery to the lysosome. Increasing evidence reveals that autophagic dysfunction is associated with human diseases, such as cancer. Paradoxically, although autophagy is well recognized as a cell survival process that promotes tumor development, it can also participate in a caspase-independent form of programmed cell death. Induction of autophagic cell death by some anticancer agents highlights the potential of this process as a cancer treatment modality. Here, we review our current understanding of the molecular mechanism of autophagy and the potential roles of autophagy in cell death, cancer development, and cancer treatment.
Collapse
Affiliation(s)
- Lin Lin
- Department of Cancer Biology; University of Massachusetts Medical School ; Worcester, MA, USA
| | - Eric H Baehrecke
- Department of Cancer Biology; University of Massachusetts Medical School ; Worcester, MA, USA
| |
Collapse
|
72
|
Farkaš R, Beňová-Liszeková D, Mentelová L, Mahmood S, Ďatková Z, Beňo M, Pečeňová L, Raška O, Šmigová J, Chase BA, Raška I, Mechler BM. Vacuole dynamics in the salivary glands ofDrosophila melanogasterduring prepupal development. Dev Growth Differ 2015; 57:74-96. [DOI: 10.1111/dgd.12193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 10/21/2014] [Accepted: 11/28/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Silvia Mahmood
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Medical Biochemistry; Jessenius Faculty of Medicine; Comenius University; Mala Hora 4 03601 Martin Slovakia
| | - Zuzana Ďatková
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Milan Beňo
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
| | - Ludmila Pečeňová
- Laboratory of Developmental Genetics; Institute of Experimental Endocrinology; Slovak Academy of Sciences; Vlárska 3 83306 Bratislava Slovakia
- Department of Genetics; Comenius University; Mlynská dolina, B-1 84215 Bratislava Slovakia
| | - Otakar Raška
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Jana Šmigová
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Bruce A. Chase
- Department of Biology; University of Nebraska at Omaha; 6001 Dodge Street Omaha NE 68182-0040 USA
| | - Ivan Raška
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
| | - Bernard M. Mechler
- Institute of Cellular Biology and Pathology; 1st Faculty of Medicine; Charles University in Prague; Albertov 4 12800 Prague Czech Republic
- German Cancer Research Centre; Neuenheimer Feld 581 D-69120 Heidelberg Germany
- VIT-University; Vellore Tamil Nadu India
| |
Collapse
|
73
|
Gautam NK, Verma P, Tapadia MG. Ecdysone regulates morphogenesis and function of Malpighian tubules in Drosophila melanogaster through EcR-B2 isoform. Dev Biol 2014; 398:163-76. [PMID: 25476260 DOI: 10.1016/j.ydbio.2014.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 10/20/2014] [Accepted: 11/11/2014] [Indexed: 10/24/2022]
Abstract
Malpighian tubules are the osmoregulatory and detoxifying organs of Drosophila and its proper development is critical for the survival of the organism. They are made up of two major cell types, the ectodermal principal cells and mesodermal stellate cells. The principal and stellate cells are structurally and physiologically distinct from each other, but coordinate together for production of isotonic fluid. Proper integration of these cells during the course of development is an important pre-requisite for the proper functioning of the tubules. We have conclusively determined an essential role of ecdysone hormone in the development and function of Malpighian tubules. Disruption of ecdysone signaling interferes with the organization of principal and stellate cells resulting in malformed tubules and early larval lethality. Abnormalities include reduction in the number of cells and the clustering of cells rather than their arrangement in characteristic wild type pattern. Organization of F-actin and β-tubulin also show aberrant distribution pattern. Malformed tubules show reduced uric acid deposition and altered expression of Na(+)/K(+)-ATPase pump. B2 isoform of ecdysone receptor is critical for the development of Malpighian tubules and is expressed from early stages of its development.
Collapse
Affiliation(s)
- Naveen Kumar Gautam
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005,Uttar Pradesh, India; Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Puja Verma
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005,Uttar Pradesh, India
| | - Madhu G Tapadia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi 221005,Uttar Pradesh, India.
| |
Collapse
|
74
|
Autophagy as a pro-death pathway. Immunol Cell Biol 2014; 93:35-42. [PMID: 25331550 DOI: 10.1038/icb.2014.85] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/01/2014] [Accepted: 09/08/2014] [Indexed: 12/12/2022]
Abstract
The evolutionarily conserved catabolic process of autophagy involves the degradation of cytoplasmic components through lysosomal enzymes. Basal levels of autophagy maintain cellular homeostasis and under stress conditions high levels of autophagy are induced. It is often under such stress conditions that high levels of autophagy and cell death have been observed, leading to the idea that autophagy may act as an executioner of cell death. However the notion of autophagy as a cell death mechanism has been controversial and remains mechanistically undefined. There is now growing evidence that in specific contexts autophagy can indeed facilitate cell death. The pro-death role of autophagy is however complicated due to the extensive cross-talk between different signalling pathways. This review summarises the examples of where autophagy acts as a means of cell death and discusses the association of autophagy with the different cell death pathways.
Collapse
|
75
|
Nelson C, Ambros V, Baehrecke EH. miR-14 regulates autophagy during developmental cell death by targeting ip3-kinase 2. Mol Cell 2014; 56:376-388. [PMID: 25306920 DOI: 10.1016/j.molcel.2014.09.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/29/2014] [Accepted: 09/03/2014] [Indexed: 12/19/2022]
Abstract
Macroautophagy (autophagy) is a lysosome-dependent degradation process that has been implicated in age-associated diseases. Autophagy is involved in both cell survival and cell death, but little is known about the mechanisms that distinguish its use during these distinct cell fates. Here, we identify the microRNA miR-14 as being both necessary and sufficient for autophagy during developmentally regulated cell death in Drosophila. Loss of miR-14 prevented induction of autophagy during salivary gland cell death, but had no effect on starvation-induced autophagy in the fat body. Moreover, misexpression of miR-14 was sufficient to prematurely induce autophagy in salivary glands, but not in the fat body. Importantly, miR-14 regulates this context-specific autophagy through its target, inositol 1,4,5-trisphosphate kinase 2 (ip3k2), thereby affecting inositol 1,4,5-trisphosphate (IP3) signaling and calcium levels during salivary gland cell death. This study provides in vivo evidence of microRNA regulation of autophagy through modulation of IP3 signaling.
Collapse
Affiliation(s)
- Charles Nelson
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Victor Ambros
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Eric H Baehrecke
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
76
|
A molecular view of autophagy in Lepidoptera. BIOMED RESEARCH INTERNATIONAL 2014; 2014:902315. [PMID: 25143951 PMCID: PMC4124216 DOI: 10.1155/2014/902315] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/06/2014] [Accepted: 06/20/2014] [Indexed: 12/17/2022]
Abstract
Metamorphosis represents a critical phase in the development of holometabolous insects, during which the larval body is completely reorganized: in fact, most of the larval organs undergo remodeling or completely degenerate before the final structure of the adult insect is rebuilt. In the past, increasing evidence emerged concerning the intervention of autophagy and apoptosis in the cell death processes that occur in larval organs of Lepidoptera during metamorphosis, but a molecular characterization of these pathways was undertaken only in recent years. In addition to developmentally programmed autophagy, there is growing interest in starvation-induced autophagy. Therefore we are now entering a new era of research on autophagy that foreshadows clarification of the role and regulatory mechanisms underlying this self-digesting process in Lepidoptera. Given that some of the most important lepidopteran species of high economic importance, such as the silkworm, Bombyx mori, belong to this insect order, we expect that this information on autophagy will be fully exploited not only in basic research but also for practical applications.
Collapse
|
77
|
Denton D, Aung-Htut MT, Lorensuhewa N, Nicolson S, Zhu W, Mills K, Cakouros D, Bergmann A, Kumar S. UTX coordinates steroid hormone-mediated autophagy and cell death. Nat Commun 2014; 4:2916. [PMID: 24336022 DOI: 10.1038/ncomms3916] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 11/12/2013] [Indexed: 02/07/2023] Open
Abstract
Correct spatial and temporal induction of numerous cell type-specific genes during development requires regulated removal of the repressive histone H3 lysine 27 trimethylation (H3K27me3) modification. Here we show that the H3K27me3 demethylase dUTX is required for hormone-mediated transcriptional regulation of apoptosis and autophagy genes during ecdysone-regulated programmed cell death of Drosophila salivary glands. We demonstrate that dUTX binds to the nuclear hormone receptor complex Ecdysone Receptor/Ultraspiracle, and is recruited to the promoters of key apoptosis and autophagy genes. Salivary gland cell death is delayed in dUTX mutants, with reduced caspase activity and autophagy that coincides with decreased apoptosis and autophagy gene transcripts. We further show that salivary gland degradation requires dUTX catalytic activity. Our findings provide evidence for an unanticipated role for UTX demethylase activity in regulating hormone-dependent cell death and demonstrate how a single transcriptional regulator can modulate a specific complex functional outcome during animal development.
Collapse
Affiliation(s)
- Donna Denton
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia [2] Division of Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - May T Aung-Htut
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Nirmal Lorensuhewa
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Shannon Nicolson
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Wenying Zhu
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Kathryn Mills
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Dimitrios Cakouros
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia
| | - Andreas Bergmann
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | - Sharad Kumar
- 1] Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, South Australia 5000, Australia [2] Division of Health Sciences, University of South Australia, Adelaide, South Australia 5001, Australia [3] Department of Medicine, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
78
|
Fernandes KM, Neves CA, Serrão JE, Martins GF. Aedes aegypti midgut remodeling during metamorphosis. Parasitol Int 2014; 63:506-12. [DOI: 10.1016/j.parint.2014.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 12/21/2013] [Accepted: 01/16/2014] [Indexed: 11/25/2022]
|
79
|
Mulakkal NC, Nagy P, Takats S, Tusco R, Juhász G, Nezis IP. Autophagy in Drosophila: from historical studies to current knowledge. BIOMED RESEARCH INTERNATIONAL 2014; 2014:273473. [PMID: 24949430 PMCID: PMC4052151 DOI: 10.1155/2014/273473] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/17/2014] [Indexed: 12/17/2022]
Abstract
The discovery of evolutionarily conserved Atg genes required for autophagy in yeast truly revolutionized this research field and made it possible to carry out functional studies on model organisms. Insects including Drosophila are classical and still popular models to study autophagy, starting from the 1960s. This review aims to summarize past achievements and our current knowledge about the role and regulation of autophagy in Drosophila, with an outlook to yeast and mammals. The basic mechanisms of autophagy in fruit fly cells appear to be quite similar to other eukaryotes, and the role that this lysosomal self-degradation process plays in Drosophila models of various diseases already made it possible to recognize certain aspects of human pathologies. Future studies in this complete animal hold great promise for the better understanding of such processes and may also help finding new research avenues for the treatment of disorders with misregulated autophagy.
Collapse
Affiliation(s)
- Nitha C. Mulakkal
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Peter Nagy
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Szabolcs Takats
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Radu Tusco
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Gábor Juhász
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Ioannis P. Nezis
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
80
|
Farkaš R, Ďatková Z, Mentelová L, Löw P, Beňová-Liszeková D, Beňo M, Sass M, Řehulka P, Řehulková H, Raška O, Kováčik L, Šmigová J, Raška I, Mechler BM. Apocrine secretion in Drosophila salivary glands: subcellular origin, dynamics, and identification of secretory proteins. PLoS One 2014; 9:e94383. [PMID: 24732043 PMCID: PMC3986406 DOI: 10.1371/journal.pone.0094383] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/14/2014] [Indexed: 01/29/2023] Open
Abstract
In contrast to the well defined mechanism of merocrine exocytosis, the mechanism of apocrine secretion, which was first described over 180 years ago, remains relatively uncharacterized. We identified apocrine secretory activity in the late prepupal salivary glands of Drosophila melanogaster just prior to the execution of programmed cell death (PCD). The excellent genetic tools available in Drosophila provide an opportunity to dissect for the first time the molecular and mechanistic aspects of this process. A prerequisite for such an analysis is to have pivotal immunohistochemical, ultrastructural, biochemical and proteomic data that fully characterize the process. Here we present data showing that the Drosophila salivary glands release all kinds of cellular proteins by an apocrine mechanism including cytoskeletal, cytosolic, mitochondrial, nuclear and nucleolar components. Surprisingly, the apocrine release of these proteins displays a temporal pattern with the sequential release of some proteins (e.g. transcription factor BR-C, tumor suppressor p127, cytoskeletal β-tubulin, non-muscle myosin) earlier than others (e.g. filamentous actin, nuclear lamin, mitochondrial pyruvate dehydrogenase). Although the apocrine release of proteins takes place just prior to the execution of an apoptotic program, the nuclear DNA is never released. Western blotting indicates that the secreted proteins remain undegraded in the lumen. Following apocrine secretion, the salivary gland cells remain quite vital, as they retain highly active transcriptional and protein synthetic activity.
Collapse
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia,
| | - Zuzana Ďatková
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia,
- Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia,
- Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Péter Löw
- Department of Anatomy and Cell Biology, Lorand Eötvös University, Budapest, Hungary
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia,
| | - Milan Beňo
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia,
| | - Miklós Sass
- Department of Anatomy and Cell Biology, Lorand Eötvös University, Budapest, Hungary
| | - Pavel Řehulka
- Institute of Molecular Pathology, Faculty of Military Health Sciences, University of Defence, Hradec Králové, Czech Republic
| | - Helena Řehulková
- 1st Department of Internal Medicine - Cardioangiology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Hradec Králové, Czech Republic
| | - Otakar Raška
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Lubomír Kováčik
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Jana Šmigová
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Ivan Raška
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Bernard M. Mechler
- Institute of Cellular Biology and Pathology, 1st Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Deutsches Krebsforschungszentrum, Heidelberg, Germany
| |
Collapse
|
81
|
Ecdysone-induced receptor tyrosine phosphatase PTP52F regulates Drosophila midgut histolysis by enhancement of autophagy and apoptosis. Mol Cell Biol 2014; 34:1594-606. [PMID: 24550005 DOI: 10.1128/mcb.01391-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The rapid removal of larval midgut is a critical developmental process directed by molting hormone ecdysone during Drosophila metamorphosis. To date, it remains unclear how the stepwise events can link the onset of ecdysone signaling to the destruction of larval midgut. This study investigated whether ecdysone-induced expression of receptor protein tyrosine phosphatase PTP52F regulates this process. The mutation of the Ptp52F gene caused significant delay in larval midgut degradation. Transitional endoplasmic reticulum ATPase (TER94), a regulator of ubiquitin proteasome system, was identified as a substrate and downstream effector of PTP52F in the ecdysone signaling. The inducible expression of PTP52F at the puparium formation stage resulted in dephosphorylation of TER94 on its Y800 residue, ensuring the rapid degradation of ubiquitylated proteins. One of the proteins targeted by dephosphorylated TER94 was found to be Drosophila inhibitor of apoptosis 1 (DIAP1), which was rapidly proteolyzed in cells with significant expression of PTP52F. Importantly, the reduced level of DIAP1 in response to inducible PTP52F was essential not only for the onset of apoptosis but also for the initiation of autophagy. This study demonstrates a novel function of PTP52F in regulating ecdysone-directed metamorphosis via enhancement of autophagic and apoptotic cell death in doomed Drosophila midguts.
Collapse
|
82
|
Boyan G, Liu Y. Timelines in the insect brain: fates of identified neural stem cells generating the central complex in the grasshopper Schistocerca gregaria. Dev Genes Evol 2014; 224:37-51. [PMID: 24343526 DOI: 10.1007/s00427-013-0462-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/02/2013] [Indexed: 11/27/2022]
Abstract
This study employs labels for cell proliferation and cell death, as well as classical histology to examine the fates of all eight neural stem cells (neuroblasts) whose progeny generate the central complex of the grasshopper brain during embryogenesis. These neuroblasts delaminate from the neuroectoderm between 25 and 30 % of embryogenesis and form a linear array running from ventral (neuroblasts Z, Y, X, and W) to dorsal (neuroblasts 1-2, 1-3, 1-4, and 1-5) along the medial border of each protocerebral hemisphere. Their stereotypic location within the array, characteristic size, and nuclear morphologies, identify these neuroblasts up to about 70 % of embryogenesis after which cell shrinkage and shape changes render progressively more cells histologically unrecognizable. Molecular labels show all neuroblasts in the array are proliferative up to 70 % of embryogenesis, but subsequently first the more ventral cells (72-75 %), and then the dorsal ones (77-80 %), cease proliferation. By contrast, neuroblasts elsewhere in the brain and optic lobe remain proliferative. Apoptosis markers label the more ventral neuroblasts first (70-72 %), then the dorsal cells (77 %), and the absence of any labeling thereafter confirms that central complex neuroblasts have exited the cell cycle via programmed cell death. Our data reveal appearance, proliferation, and cell death proceeding as successive waves from ventral to dorsal along the array of neuroblasts. The resulting timelines offer a temporal blueprint for building the neuroarchitecture of the various modules of the central complex.
Collapse
Affiliation(s)
- George Boyan
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstrasse 2, 82152, Planegg-Martinsried, Germany,
| | | |
Collapse
|
83
|
Neuman SD, Ihry RJ, Gruetzmacher KM, Bashirullah A. INO80-dependent regression of ecdysone-induced transcriptional responses regulates developmental timing in Drosophila. Dev Biol 2014; 387:229-39. [PMID: 24468295 DOI: 10.1016/j.ydbio.2014.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 01/11/2014] [Indexed: 11/19/2022]
Abstract
Sequential pulses of the steroid hormone ecdysone regulate the major developmental transitions in Drosophila, and the duration of each developmental stage is determined by the length of time between ecdysone pulses. Ecdysone regulates biological responses by directly initiating target gene transcription. In turn, these transcriptional responses are known to be self-limiting, with mechanisms in place to ensure regression of hormone-dependent transcription. However, the biological significance of these transcriptional repression mechanisms remains unclear. Here we show that the chromatin remodeling protein INO80 facilitates transcriptional repression of ecdysone-regulated genes during prepupal development. In ino80 mutant animals, inefficient repression of transcriptional responses to the late larval ecdysone pulse delays the onset of the subsequent prepupal ecdysone pulse, resulting in a significantly longer prepupal stage. Conversely, increased expression of ino80 is sufficient to shorten the prepupal stage by increasing the rate of transcriptional repression. Furthermore, we demonstrate that enhancing the rate of regression of the mid-prepupal competence factor βFTZ-F1 is sufficient to determine the timing of head eversion and thus the duration of prepupal development. Although ino80 is conserved from yeast to humans, this study represents the first characterization of a bona fide ino80 mutation in any metazoan, raising the possibility that the functions of ino80 in transcriptional repression and developmental timing are evolutionarily conserved.
Collapse
Affiliation(s)
- Sarah D Neuman
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Robert J Ihry
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kelly M Gruetzmacher
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA; College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Arash Bashirullah
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA; Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
84
|
Abstract
Steroid hormones trigger a wide variety of biological responses through stage- and tissue-specific activation of target gene expression. The mechanisms that provide specificity to systemically released pulses of steroids, however, remain poorly understood. We previously completed a forward genetic screen for mutations that disrupt the destruction of larval salivary glands during metamorphosis in Drosophila melanogaster, a process triggered by the steroid hormone 20-hydroxyecdysone (ecdysone). Here, we characterize 10 complementation groups mapped to genes from this screen. Most of these mutations disrupt the ecdysone-induced expression of death activators, thereby failing to initiate tissue destruction. However, other responses to ecdysone, even within salivary glands, occur normally in mutant animals. Many of these newly identified regulators of ecdysone signaling, including brwd3, med12, med24, pak, and psg2, represent novel components of the ecdysone-triggered transcriptional hierarchy. These genes function combinatorially to provide specificity to ecdysone pulses, amplifying the hormonal cue in a stage-, tissue-, and target gene-specific manner. Most of the ecdysone response genes identified in this screen encode homologs of mammalian nuclear receptor coregulators, demonstrating an unexpected degree of functional conservation in the mechanisms that regulate steroid signaling between insects and mammals.
Collapse
|
85
|
Tasdemir-Yilmaz OE, Freeman MR. Astrocytes engage unique molecular programs to engulf pruned neuronal debris from distinct subsets of neurons. Genes Dev 2013; 28:20-33. [PMID: 24361692 PMCID: PMC3894410 DOI: 10.1101/gad.229518.113] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neural circuit assembly involves an initial overproduction of neurons and synaptic connections, followed by their selective elimination. Tasdemir-Yilmaz and Freeman show that Drosophila larval astrocytes are the primary phagocytic cell type in the pupal CNS neuropil. MB γ neuron axons are engulfed by astrocytes using the Draper and Crk/Mbc/dCed-12 signaling pathways, while Crk/Mbc/dCed-12, but not Draper, mediates the elimination of vCrz+ neurites. Eliminating Draper signaling delays early vCrz+ neurite degeneration, suggesting that glial cells promote neurite destruction through engulfment signaling. Precise neural circuit assembly is achieved by initial overproduction of neurons and synapses, followed by refinement through elimination of exuberant neurons and synapses. Glial cells are the primary cells responsible for clearing neuronal debris, but the cellular and molecular basis of glial pruning is poorly defined. Here we show that Drosophila larval astrocytes transform into phagocytes through activation of a cell-autonomous, steroid-dependent program at the initiation of metamorphosis and are the primary phagocytic cell type in the pupal neuropil. We examined the developmental elimination of two neuron populations—mushroom body (MB) γ neurons and vCrz+ neurons (expressing Corazonin [Crz] neuropeptide in the ventral nerve cord [VNC])—where only neurites are pruned or entire cells are eliminated, respectively. We found that MB γ axons are engulfed by astrocytes using the Draper and Crk/Mbc/dCed-12 signaling pathways in a partially redundant manner. In contrast, while elimination of vCrz+ cell bodies requires Draper, elimination of vCrz+ neurites is mediated by Crk/Mbc/dCed-12 but not Draper. Intriguingly, we also found that elimination of Draper delayed vCrz+ neurite degeneration, suggesting that glia promote neurite destruction through engulfment signaling. This study identifies a novel role for astrocytes in the clearance of synaptic and neuronal debris and for Crk/Mbc/dCed-12 as a new glial pathway mediating pruning and reveals, unexpectedly, that the engulfment signaling pathways engaged by glia depend on whether neuronal debris was generated through cell death or local pruning.
Collapse
Affiliation(s)
- Ozge E Tasdemir-Yilmaz
- Department of Neurobiology, Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
86
|
Denton D, Aung-Htut MT, Kumar S. Developmentally programmed cell death in Drosophila. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3499-3506. [DOI: 10.1016/j.bbamcr.2013.06.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/16/2013] [Indexed: 12/24/2022]
|
87
|
Hassel C, Zhang B, Dixon M, Calvi BR. Induction of endocycles represses apoptosis independently of differentiation and predisposes cells to genome instability. Development 2013; 141:112-23. [PMID: 24284207 DOI: 10.1242/dev.098871] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The endocycle is a common developmental cell cycle variation wherein cells become polyploid through repeated genome duplication without mitosis. We previously showed that Drosophila endocycling cells repress the apoptotic cell death response to genotoxic stress. Here, we investigate whether it is differentiation or endocycle remodeling that promotes apoptotic repression. We find that when nurse and follicle cells switch into endocycles during oogenesis they repress the apoptotic response to DNA damage caused by ionizing radiation, and that this repression has been conserved in the genus Drosophila over 40 million years of evolution. Follicle cells defective for Notch signaling failed to switch into endocycles or differentiate and remained apoptotic competent. However, genetic ablation of mitosis by knockdown of Cyclin A or overexpression of fzr/Cdh1 induced follicle cell endocycles and repressed apoptosis independently of Notch signaling and differentiation. Cells recovering from these induced endocycles regained apoptotic competence, showing that repression is reversible. Recovery from fzr/Cdh1 overexpression also resulted in an error-prone mitosis with amplified centrosomes and high levels of chromosome loss and fragmentation. Our results reveal an unanticipated link between endocycles and the repression of apoptosis, with broader implications for how endocycles may contribute to genome instability and oncogenesis.
Collapse
Affiliation(s)
- Christiane Hassel
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | | | | |
Collapse
|
88
|
Guan R, El-Rass S, Spillane D, Lam S, Wang Y, Wu J, Chen Z, Wang A, Jia Z, Keating A, Hu J, Wen XY. rbm47, a novel RNA binding protein, regulates zebrafish head development. Dev Dyn 2013; 242:1395-404. [PMID: 24038582 DOI: 10.1002/dvdy.24039] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 08/02/2013] [Accepted: 08/14/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Vertebrate trunk induction requires inhibition of bone morphogenetic protein (BMP) signaling, whereas vertebrate head induction requires concerted inhibition of both Wnt and BMP signaling. RNA binding proteins play diverse roles in embryonic development and their roles in vertebrate head development remain to be elucidated. RESULTS We first characterized the human RBM47 as an RNA binding protein that specifically binds RNA but not single-stranded DNA. Next, we knocked down rbm47 gene function in zebrafish using morpholinos targeting the start codon and exon-1/intron-1 splice junction. Down-regulation of rbm47 resulted in headless and small head phenotypes, which can be rescued by a wnt8a blocking morpholino. To further reveal the mechanism of rbm47's role in head development, microarrays were performed to screen genes differentially expressed in normal and knockdown embryos. epcam and a2ml were identified as the most significantly up- and down-regulated genes, respectively. The microarrays also confirmed up-regulation of several genes involved in head development, including gsk3a, otx2, and chordin, which are important regulators of Wnt signaling. CONCLUSIONS Altogether, our findings reveal that Rbm47 is a novel RNA-binding protein critical for head formation and embryonic patterning during zebrafish embryogenesis which may act through a Wnt8a signaling pathway.
Collapse
Affiliation(s)
- Rui Guan
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Medicine, Physiology and Institute of Medical Science, University of Toronto, Canada; Department of Laboratory Medicine and Pathology, University of Toronto & Program in Physiology and Experimental Medicine, The Hospital for Sick Children, Toronto, Canada; Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital and Cancer Research Institute, Central South, Hunan Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Zirin J, Cheng D, Dhanyasi N, Cho J, Dura JM, Vijayraghavan K, Perrimon N. Ecdysone signaling at metamorphosis triggers apoptosis of Drosophila abdominal muscles. Dev Biol 2013; 383:275-84. [PMID: 24051228 DOI: 10.1016/j.ydbio.2013.08.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/12/2013] [Accepted: 08/19/2013] [Indexed: 11/28/2022]
Abstract
One of the most dramatic examples of programmed cell death occurs during Drosophila metamorphosis, when most of the larval tissues are destroyed in a process termed histolysis. Much of our understanding of this process comes from analyses of salivary gland and midgut cell death. In contrast, relatively little is known about the degradation of the larval musculature. Here, we analyze the programmed destruction of the abdominal dorsal exterior oblique muscle (DEOM) which occurs during the first 24h of metamorphosis. We find that ecdysone signaling through Ecdysone receptor isoform B1 is required cell autonomously for the muscle death. Furthermore, we show that the orphan nuclear receptor FTZ-F1, opposed by another nuclear receptor, HR39, plays a critical role in the timing of DEOM histolysis. Finally, we show that unlike the histolysis of salivary gland and midgut, abdominal muscle death occurs by apoptosis, and does not require autophagy. Thus, there is no set rule as to the role of autophagy and apoptosis during Drosophila histolysis.
Collapse
|
90
|
Abstract
Many organs respond to physiological challenges by changing tissue size or composition. Such changes may originate from tissue-specific stem cells and their supportive environment (niche). The endocrine system is a major effector and conveyor of physiological changes and as such could alter stem cell behavior in various ways. In this review, we examine how hormones affect stem cell biology in four different organs: the ovary, intestine, hematopoietic system, and mammary gland. Hormones control every stage of stem cell life, including establishment, expansion, maintenance, and differentiation. The effects can be cell autonomous or non-cell autonomous through the niche. Moreover, a single hormone can affect different stem cells in different ways or affect the same stem cell differently at various developmental times. The vast complexity and diversity of stem cell responses to hormonal cues allow hormones to coordinate the body's reaction to physiological challenges.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100 Israel; ,
| | | |
Collapse
|
91
|
Uba1 functions in Atg7- and Atg3-independent autophagy. Nat Cell Biol 2013; 15:1067-78. [PMID: 23873149 PMCID: PMC3762904 DOI: 10.1038/ncb2804] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
Abstract
Autophagy is a conserved process that delivers components of the cytoplasm to lysosomes for degradation. The E1 and E2 enzymes encoded by Atg7 and Atg3 are thought to be essential for autophagy involving the ubiquitin-like protein Atg8. Here, we describe an Atg7- and Atg3-independent autophagy pathway that facilitates programmed reduction of cell size during intestine cell death. Although multiple components of the core autophagy pathways, including Atg8, are required for autophagy and cells to shrink in the midgut of the intestine, loss of either Atg7 or Atg3 function does not influence these cellular processes. Rather, Uba1, the E1 used in ubiquitination, is required for autophagy and reduction of cell size. Our data reveal that distinct autophagy programs are used by different cells within an animal, and disclose an unappreciated role for ubiquitin activation in autophagy.
Collapse
|
92
|
Liu Y, Boyan G. Glia associated with central complex lineages in the embryonic brain of the grasshopper Schistocerca gregaria. Dev Genes Evol 2013; 223:213-23. [PMID: 23494665 DOI: 10.1007/s00427-013-0439-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/27/2013] [Indexed: 12/17/2022]
Abstract
We have investigated the pattern of glia associated with central complex lineages in the embryonic brain of the grasshopper Schistocerca gregaria. Using the glia-specific marker Repo, we identified glia associated externally with such lineages, termed lineage-extrinsic glia, and glia located internally within the lineages, termed lineage-intrinsic glia. Populations of both glial types increase up to 60 % of embryogenesis, and thereafter decrease. Extrinsic glia change their locations over time, while intrinsic ones are consistently found in the more apical part of a lineage. Apoptosis is not observed for either glial type, suggesting migration is a likely mechanism accounting for changes in glial number. Proliferative glia are present both within and without individual lineages and two glial clusters associated with the lineages, one apically and the other basally, may represent sources of glia.
Collapse
Affiliation(s)
- Yu Liu
- Developmental Neurobiology Group, Biocenter, Ludwig-Maximilians-Universität, Grosshadernerstr. 2, 82152, Martinsried, Germany.
| | | |
Collapse
|
93
|
Ecdysone-dependent and ecdysone-independent programmed cell death in the developing optic lobe of Drosophila. Dev Biol 2013; 374:127-41. [DOI: 10.1016/j.ydbio.2012.11.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/30/2012] [Accepted: 11/02/2012] [Indexed: 12/14/2022]
|
94
|
Effects of diet and development on the Drosophila lipidome. Mol Syst Biol 2013; 8:600. [PMID: 22864382 PMCID: PMC3421444 DOI: 10.1038/msb.2012.29] [Citation(s) in RCA: 210] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 06/25/2012] [Indexed: 12/12/2022] Open
Abstract
Cells produce tens of thousands of different lipid species, but the importance of this complexity in vivo is unclear. Analysis of individual tissues and cell types has revealed differences in abundance of individual lipid species, but there has been no comprehensive study comparing tissue lipidomes within a single developing organism. Here, we used quantitative shotgun profiling by high-resolution mass spectrometry to determine the absolute (molar) content of 250 species of 14 major lipid classes in 6 tissues of animals at 27 developmental stages raised on 4 different diets. Comparing these lipidomes revealed unexpected insights into lipid metabolism. Surprisingly, the fatty acids present in dietary lipids directly influence tissue phospholipid composition throughout the animal. Furthermore, Drosophila differentially regulates uptake, mobilization and tissue accumulation of specific sterols, and undergoes unsuspected shifts in fat metabolism during larval and pupal development. Finally, we observed striking differences between tissue lipidomes that are conserved between phyla. This study provides a comprehensive, quantitative and expandable resource for further pharmacological and genetic studies of metabolic disorders and molecular mechanisms underlying dietary response.
Collapse
|
95
|
Abstract
Macroautophagy (autophagy) is a conserved catabolic process that targets cytoplasmic components to lysosomes for degradation. Autophagy is required for cellular homeostasis and cell survival in response to starvation and stress, and paradoxically, it also plays a role in programmed cell death during development. The mechanisms that regulate the relationship between autophagy, cell survival, and cell death are poorly understood. Here we review research in Drosophila that has provided insights into the regulation of autophagy by steroid hormones and nutrient restriction and discuss how autophagy influences cell growth, nutrient utilization, cell survival, and cell death.
Collapse
|
96
|
Zeng X, Hou SX. Broad relays hormone signals to regulate stem cell differentiation in Drosophila midgut during metamorphosis. Development 2012; 139:3917-25. [PMID: 23048182 DOI: 10.1242/dev.083030] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Like the mammalian intestine, the Drosophila adult midgut is constantly replenished by multipotent intestinal stem cells (ISCs). Although it is well known that adult ISCs arise from adult midgut progenitors (AMPs), relatively little is known about the mechanisms that regulate AMP specification. Here, we demonstrate that Broad (Br)-mediated hormone signaling regulates AMP specification. Br is highly expressed in AMPs temporally during the larva-pupa transition stage, and br loss of function blocks AMP differentiation. Furthermore, Br is required for AMPs to develop into functional ISCs. Conversely, br overexpression drives AMPs toward premature differentiation. In addition, we found that Br and Notch (N) signaling function in parallel pathways to regulate AMP differentiation. Our results reveal a molecular mechanism whereby Br-mediated hormone signaling directly regulates stem cells to generate adult cells during metamorphosis.
Collapse
Affiliation(s)
- Xiankun Zeng
- The Mouse Cancer Genetics Program, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, USA.
| | | |
Collapse
|
97
|
Kim MY, Song HY, Kim JH, Kim BY, Park SW, Sung DK, Park HH, Jeon SH, Chung IS, Lee BH. Silkworm 30K protein inhibits ecdysone-induced apoptosis by blocking the binding of ultraspiracle to ecdysone receptor-B1 in cultured Bm5 cells. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2012; 81:136-147. [PMID: 22890884 DOI: 10.1002/arch.21050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study investigates the mechanism through which increased 30K protein inhibits ecdysone-induced apoptosis in the Bm5 silkworm ovarian cell line. Treatment of Bm5 cells with 20-hydroxyecdysone (20E) after transfection with the pIZT/V5-His control vector triggered apoptosis, but 20E treatment did not trigger apoptosis in Bm5 cells transfected with the pIZT/30K/V5-His vector. To confirm its inhibitory effect on apoptosis, 30K protein was first purified from Escherichia coli transformed with a 30K expression vector and used to generate specific antibodies in mice. Anti-30K antiserum was used to confirm synthesis of the 30K protein in pIZT/30K/V5-His-transfected Bm5 cells and to detect 30K protein binding to the ecdysone receptor-B1 (EcR-B1). Anti-30K antiserum was used to immunoprecipitate protein complexes containing 30K from Bm5 cells transfected with pIZT/30K/V5-His vector and treated with 20E. We observed that 30K proteins bound primarily to the EcR-B1 and not to ultraspiracle (USP). Reciprocal immunoprecipitation of EcR-B1-containing complexes from Bm5 cells transfected with control pIZT/V5-His vector and treated with 20E showed that EcR-B1 bound to USP in the absence of 30K but did not bind to USP in pIZT/30K/V5-His-transfected Bm5 cells. These results demonstrate that 30K proteins block USP binding to EcR-B1 through formation of a 30K/EcR-B1 complex, resulting in inhibition of 20E-induced Bm5 cell apoptosis.
Collapse
Affiliation(s)
- Mi Young Kim
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Togane Y, Ayukawa R, Hara Y, Akagawa H, Iwabuchi K, Tsujimura H. Spatio-temporal pattern of programmed cell death in the developing Drosophila optic lobe. Dev Growth Differ 2012; 54:503-18. [PMID: 22587328 DOI: 10.1111/j.1440-169x.2012.01340.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A large number of cells die via programmed cell death during the normal development of the Drosophila optic lobe. In this study, we report the precise spatial and temporal pattern of cell death in this organ. Cell death in the developing optic lobe occurs in two distinct phases. The first phase extends from the start of metamorphosis to the mid-pupal stage. During this phase, a large number of cells die in the optic lobe as a whole, with a peak of cell death at an early pupal stage in the lamina and medulla cortices and the region of the T2/T3/C neurons, and a smaller number of dead cells observed in the lobula plate cortex. The second phase extends from the mid-pupal stage to eclosion. Throughout this period, a small number of dying cells can be observed, with a small peak at a late pupal stage. Most of the dying cells are neurons. During the first phase, dying cells are distributed in specific patterns in cortices. The lamina cortex contains two distinct clusters of dying cells; the medulla cortex, four clusters; the lobula plate cortex, one cluster; and the region of the T2/T3/C neurons, one cluster. Many of the clusters maintain their distinct positions in the optic lobe but others extend the region they cover during development. The presence of distinct clusters of dying cells at different phases suggests that distinct mechanisms control cell death during different stages of optic lobe development in Drosophila.
Collapse
Affiliation(s)
- Yu Togane
- Developmental Biology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-si, Tokyo, 183-8509, Japan
| | | | | | | | | | | |
Collapse
|
99
|
Manaboon M, Yasanga T, Sakurai S, Singtripop T. Programmed cell death of larval tissues induced by juvenile hormone in the bamboo borer, Omphisa fuscidentalis. JOURNAL OF INSECT PHYSIOLOGY 2012; 58:1202-1208. [PMID: 22732232 DOI: 10.1016/j.jinsphys.2012.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/31/2012] [Accepted: 06/05/2012] [Indexed: 06/01/2023]
Abstract
Programmed cell death (PCD) plays a critical role during animal development through the destruction of unneeded cells and tissues. In some insects, the prothoracic glands (PGs) and anterior silk glands (ASGs) are larval-specific tissues that are normally eliminated by PCD after pupation. Previous studies report that juvenile hormone analog (JHA) terminates the larval diapause of Omphisa fuscidentalis by increasing the hemolymph ecdysteroids that trigger PCD. Because JHA may indirectly induce the PCD of the PGs and ASGs of Omphisa diapausing larvae, the effects of JHA on the induction of PCD were determined. The application of 1μg JHA induced PCD in the PGs and ASGs of larvae identified as stage G0 (prior to pupation). The injection of 1μg 20E triggered the PCD of the ASGs when the larvae expressed a G0-G1 morphology, whereas PCD occurred in the PGs on day 1 post-injection. Histological studies revealed similar patterns of morphological changes during the PG and ASG PCD in the JHA- and 20E-treated larvae. Furthermore, to confirm that PCD was induced by a high ecdysteroid level that increases after JHA application, the expression profiles of EcR-A and EcR-B1 in the PGs and ASGs from the JHA-treated larvae were examined, and the results showed that the expression levels of EcR-A and EcR-B1 mRNA increased during the G0 stage. These results suggest that JHA may be involved in PCD by increasing the ecdysteroid titer, leading to termination of the larval diapause period in Omphisa fuscidentalis.
Collapse
Affiliation(s)
- Manaporn Manaboon
- Endocrinology Research Laboratory, Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| | | | | | | |
Collapse
|
100
|
Immune response and anti-microbial peptides expression in Malpighian tubules of Drosophila melanogaster is under developmental regulation. PLoS One 2012; 7:e40714. [PMID: 22808242 PMCID: PMC3395640 DOI: 10.1371/journal.pone.0040714] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 06/12/2012] [Indexed: 12/19/2022] Open
Abstract
Malpighian tubules (MT) of Drosophila melanogaster are osmoregulatory organs that maintain the ionic balance and remove toxic substances from the body. Additionally they act as autonomous immune sensing organs, which secrete antimicrobial peptides in response to invading microbial pathogens. We show that the antimicrobial peptides (AMP) diptericin, cecropinA, drosocin and attacinA are constitutively expressed and are regulated in developmental stage specific manner. Their developmental expression begins from 3rd instar larval stage and an immune challenge increases the expression several folds. Spatial variatons in the level of expression along the MT tissue are observed. The mortality of 3rd instar larvae fed on bacterial food is much less than that of the earlier larval stages, coinciding with the onset of innate immunity response in MT. Ectopic expression of AMP imparts better resistance to infection while, loss of function of one of the AMP through directed RNAi reduces host survival after immune challenge. The AMP secreted from the MT exhibit bactericidal activity. Expression of the NF-κB transcription factor, Relish, also coincides with activation of immune responsive genes in MT, demonstrating that immune regulation in MT is under developmental control and is governed by the Imd pathway.
Collapse
|