1
|
Cao Y, Xu L, Xiong X, Liu X. Expansion and diversity of caspases in Mytilus coruscus contribute to larval metamorphosis and environmental adaptation. BMC Genomics 2024; 25:314. [PMID: 38532358 DOI: 10.1186/s12864-024-10238-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Apoptosis is involved (directly and indirectly) in several physiological processes including tissue remodeling during the development, the turnover of immune cells, and a defense against harmful stimuli. The disordered apoptotic process participates in the pathogenesis of various diseases, such as neoplasms, and chronic inflammatory or systemic autoimmune diseases, which are associated with its inadequate regulation. Caspases are vital components of the apoptotic pathway that are involved in developmental and immune processes. However, genome-wide identification and functional analysis of caspase have not been conducted in Mytilus coruscus, which is an economically important bivalve. RESULTS Here, 47 caspase genes were identified from the genomes of M. coruscus, and the expansion of caspase-2/9 and caspase-3/6/7 genes were observed. Tandem duplication acts as an essential driver of gene expansion. The expanded caspase genes were highly diverse in terms of sequence, domain structure, and spatiotemporal expression profiles, suggesting their functional differentiation. The high expression of the expanded caspase genes at the pediveliger larvae stage and the result of apoptosis location in the velum suggest that the apoptosis mediated by them plays a critical role in the metamorphosis of M. coruscus larvae. In gill, caspase genes respond differently to the challenge of different strains, and most caspase-2/9 and caspase-3/6/7 genes were induced by copper stress, whereas caspase-8/10 genes were suppressed. Additionally, most caspase genes were upregulated in the mantle under ocean acidification which could weaken the biomineralization capacity of the mantle tissue. CONCLUSIONS These results provide a comprehensive overview of the evolution and function of the caspase family and enhanced the understanding of the biological function of caspases in M. coruscus larval development and response to biotic and abiotic challenges.
Collapse
Affiliation(s)
- Yanfei Cao
- National Engineering Research Center For Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - Linxiang Xu
- National Engineering Research Center For Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - Xinwei Xiong
- National Engineering Research Center For Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - Xiao Liu
- National Engineering Research Center For Marine Aquaculture, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China.
| |
Collapse
|
2
|
Wei Z, Ding W, Li M, Shi J, Wang H, Wang Y, Li Y, Xu Y, Hu J, Bao Z, Hu X. The Caspase Homologues in Scallop Chlamys farreri and Their Expression Responses to Toxic Dinoflagellates Exposure. Toxins (Basel) 2022; 14:toxins14020108. [PMID: 35202135 PMCID: PMC8878197 DOI: 10.3390/toxins14020108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
The cysteine aspartic acid-specific protease (caspase) family is distributed across vertebrates and invertebrates, and its members are involved in apoptosis and response to cellular stress. The Zhikong scallop (Chlamys farreri) is a bivalve mollusc that is well adapted to complex marine environments, yet the diversity of caspase homologues and their expression patterns in the Zhikong scallop remain largely unknown. Here, we identified 30 caspase homologues in the genome of the Zhikong scallop and analysed their expression dynamics during all developmental stages and following exposure to paralytic shellfish toxins (PSTs). The 30 caspase homologues were classified as initiators (caspases-2/9 and caspases-8/10) or executioners (caspases-3/6/7 and caspases-3/6/7-like) and displayed increased copy numbers compared to those in vertebrates. Almost all of the caspase-2/9 genes were highly expressed throughout all developmental stages from zygote to juvenile, and their expression in the digestive gland and kidney was slightly influenced by PSTs. The caspase-8/10 genes were highly expressed in the digestive gland and kidney, while PSTs inhibited their expression in these two organs. After exposure to different Alexandrium PST-producing algae (AM-1 and ACDH), the number of significantly up-regulated caspase homologues in the digestive gland increased with the toxicity level of PST derivatives, which might be due to the higher toxicity of GTXs produced by AM-1 compared to the N-sulphocarbamoyl analogues produced by ACDH. However, the effect of these two PST-producing algae strains on caspase expression in the kidney seemed to be stronger, possibly because the PST derivatives were transformed into highly toxic compounds in scallop kidney, and suggested an organ-dependent response to PSTs. These results indicate the dedicated control of caspase gene expression and highlight their contribution to PSTs in C. farreri. This work provides a further understanding of the role of caspase homologues in the Zhikong scallop and can guide future studies focussing on the role of caspases and their interactions with PSTs.
Collapse
Affiliation(s)
- Zhongcheng Wei
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Wei Ding
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Moli Li
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Jiaoxia Shi
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Huizhen Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (H.W.); (X.H.); Tel.: +86-0532-8203-1970 (X.H.)
| | - Yangrui Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Yubo Li
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Yiqiang Xu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
| | - Jingjie Hu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Laboratory of Tropical Marine Germplasm Resources and Breeding Engineering, Sanya Oceanographic Institution, Ocean University of China, Sanya 572000, China
| | - Xiaoli Hu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Z.W.); (W.D.); (M.L.); (J.S.); (Y.W.); (Y.L.); (Y.X.); (J.H.); (Z.B.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (H.W.); (X.H.); Tel.: +86-0532-8203-1970 (X.H.)
| |
Collapse
|
3
|
Verghese S, Su TT. Ionizing radiation induces stem cell-like properties in a caspase-dependent manner in Drosophila. PLoS Genet 2018; 14:e1007659. [PMID: 30462636 PMCID: PMC6248896 DOI: 10.1371/journal.pgen.1007659] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/27/2018] [Indexed: 11/18/2022] Open
Abstract
Cancer treatments including ionizing radiation (IR) can induce cancer stem cell-like properties in non-stem cancer cells, an outcome that can interfere with therapeutic success. Yet, we understand little about what consequences of IR induces stem cell like properties and why some cancer cells show this response but not others. In previous studies, we identified a pool of epithelial cells in Drosophila larval wing discs that display IR-induced stem cell-like properties. These cells are resistant to killing by IR and, after radiation damage, change fate and translocate to regenerate parts of the disc that suffered more cell death. Here, we report the identification of two new pools of cells with IR-induced regenerative capability. We addressed how IR exposure results in the induction of stem cell-like behavior, and found a requirement for IR-induced caspase activity and for Zfh2, a transcription factor and an effector in the JAK/STAT pathway. Unexpectedly, the requirement for caspase activity was cell-autonomous within cell populations that display regenerative behavior. We propose a model in which the requirement for caspase activity and Zfh2 can be explained by apoptotic and non-apoptotic functions of caspases in the induction of stem cell-like behavior. Ionizing Radiation (IR), alone or in combination with other therapies, is used to treat an estimated half of all cancer patients. Yet, we understand little about why some tumors cells respond to treatment while others grow back (regenerate). We identified specific pools of cells within a Drosophila organ that are capable of regeneration after damage by IR. We also identified what it is about IR damage that allows these cells to regenerate. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States of America
| | - Tin Tin Su
- University of Colorado Comprehensive Cancer Center, Anschutz Medical Campus, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
4
|
Vishal K, Bawa S, Brooks D, Bauman K, Geisbrecht ER. Thin is required for cell death in the Drosophila abdominal muscles by targeting DIAP1. Cell Death Dis 2018; 9:740. [PMID: 29970915 PMCID: PMC6030163 DOI: 10.1038/s41419-018-0756-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/28/2018] [Accepted: 05/30/2018] [Indexed: 12/23/2022]
Abstract
In holometabolous insects, developmentally controlled programmed cell death (PCD) is a conserved process that destroys a subset of larval tissues for the eventual creation of new adult structures. This process of histolysis is relatively well studied in salivary gland and midgut tissues, while knowledge concerning larval muscle destruction is limited. Here, we have examined the histolysis of a group of Drosophila larval abdominal muscles called the dorsal external oblique muscles (DEOMs). Previous studies have defined apoptosis as the primary mediator of DEOM breakdown, whose timing is controlled by ecdysone signaling. However, very little is known about other factors that contribute to DEOM destruction. In this paper, we examine the role of thin (tn), which encodes for the Drosophila homolog of mammalian TRIM32, in the regulation of DEOM histolysis. We find that loss of Tn blocks DEOM degradation independent of ecdysone signaling. Instead, tn genetically functions in a pathway with the death-associated inhibitor of apoptosis (DIAP1), Dronc, and death-associated APAF1-related killer (Dark) to regulate apoptosis. Importantly, blocking Tn results in the absence of active Caspase-3 immunostaining, upregulation of DIAP1 protein levels, and inhibition of Dronc activation. DIAP1 and Dronc mRNA levels are not altered in tn mutants, showing that Tn acts post-transcriptionally on DIAP1 to regulate apoptosis. Herein, we also find that the RING domain of Tn is required for DEOM histolysis as loss of this domain results in higher DIAP1 levels. Together, our results suggest that the direct control of DIAP1 levels, likely through the E3 ubiquitin ligase activity of Tn, provides a mechanism to regulate caspase activity and to facilitate muscle cell death.
Collapse
Affiliation(s)
- Kumar Vishal
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - David Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Kenneth Bauman
- Department of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas, Kansas City, MO, 64110, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
5
|
Apoptotic mechanisms during competition of ribosomal protein mutant cells: roles of the initiator caspases Dronc and Dream/Strica. Cell Death Differ 2015; 22:1300-12. [PMID: 25613379 DOI: 10.1038/cdd.2014.218] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 10/11/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
Abstract
Heterozygosity for mutations in ribosomal protein genes frequently leads to a dominant phenotype of retarded growth and small adult bristles in Drosophila (the Minute phenotype). Cells with Minute genotypes are subject to cell competition, characterized by their selective apoptosis and removal in mosaic tissues that contain wild-type cells. Competitive apoptosis was found to depend on the pro-apoptotic reaper, grim and head involution defective genes but was independent of p53. Rp/+ cells are protected by anti-apoptotic baculovirus p35 expression but lacked the usual hallmarks of 'undead' cells. They lacked Dronc activity, and neither expression of dominant-negative Dronc nor dronc knockdown by dsRNA prevented competitive apoptosis, which also continued in dronc null mutant cells or in the absence of the initiator caspases dredd and dream/strica. Only simultaneous knockdown of dronc and dream/strica by dsRNA was sufficient to protect Rp/+ cells from competition. By contrast, Rp/Rp cells were also protected by baculovirus p35, but Rp/Rp death was dronc-dependent, and undead Rp/Rp cells exhibited typical dronc-dependent expression of Wingless. Independence of p53 and unusual dependence on Dream/Strica distinguish competitive cell death from noncompetitive apoptosis of Rp/Rp cells and from many other examples of cell death.
Collapse
|
6
|
Proteomic survey reveals altered energetic patterns and metabolic failure prior to retinal degeneration. J Neurosci 2014; 34:2797-812. [PMID: 24553922 DOI: 10.1523/jneurosci.2982-13.2014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inherited mutations that lead to misfolding of the visual pigment rhodopsin (Rho) are a prominent cause of photoreceptor neuron (PN) degeneration and blindness. How Rho proteotoxic stress progressively impairs PN viability remains unknown. To identify the pathways that mediate Rho toxicity in PNs, we performed a comprehensive proteomic profiling of retinas from Drosophila transgenics expressing Rh1(P37H), the equivalent of mammalian Rho(P23H), the most common Rho mutation linked to blindness in humans. Profiling of young Rh1(P37H) retinas revealed a coordinated upregulation of energy-producing pathways and attenuation of energy-consuming pathways involving target of rapamycin (TOR) signaling, which was reversed in older retinas at the onset of PN degeneration. We probed the relevance of these metabolic changes to PN survival by using a combination of pharmacological and genetic approaches. Chronic suppression of TOR signaling, using the inhibitor rapamycin, strongly mitigated PN degeneration, indicating that TOR signaling activation by chronic Rh1(P37H) proteotoxic stress is deleterious for PNs. Genetic inactivation of the endoplasmic reticulum stress-induced JNK/TRAF1 axis as well as the APAF-1/caspase-9 axis, activated by damaged mitochondria, dramatically suppressed Rh1(P37H)-induced PN degeneration, identifying the mitochondria as novel mediators of Rh1(P37H) toxicity. We thus propose that chronic Rh1(P37H) proteotoxic stress distorts the energetic profile of PNs leading to metabolic imbalance, mitochondrial failure, and PN degeneration and therapies normalizing metabolic function might be used to alleviate Rh1(P37H) toxicity in the retina. Our study offers a glimpse into the intricate higher order interactions that underlie PN dysfunction and provides a useful resource for identifying other molecular networks that mediate Rho toxicity in PNs.
Collapse
|
7
|
Male-killing Spiroplasma induces sex-specific cell death via host apoptotic pathway. PLoS Pathog 2014; 10:e1003956. [PMID: 24550732 PMCID: PMC3923752 DOI: 10.1371/journal.ppat.1003956] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 01/12/2014] [Indexed: 11/19/2022] Open
Abstract
Some symbiotic bacteria cause remarkable reproductive phenotypes like cytoplasmic incompatibility and male-killing in their host insects. Molecular and cellular mechanisms underlying these symbiont-induced reproductive pathologies are of great interest but poorly understood. In this study, Drosophila melanogaster and its native Spiroplasma symbiont strain MSRO were investigated as to how the host's molecular, cellular and morphogenetic pathways are involved in the symbiont-induced male-killing during embryogenesis. TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, anti-cleaved-Caspase-3 antibody staining, and apoptosis-deficient mutant analysis unequivocally demonstrated that the host's apoptotic pathway is involved in Spiroplasma-induced male-specific embryonic cell death. Double-staining with TUNEL and an antibody recognizing epidermal marker showed that embryonic epithelium is the main target of Spiroplasma-induced male-specific apoptosis. Immunostaining with antibodies against markers of differentiated and precursor neural cells visualized severe neural defects specifically in Spiroplasma-infected male embryos as reported in previous studies. However, few TUNEL signals were detected in the degenerate nervous tissues of male embryos, and the Spiroplasma-induced neural defects in male embryos were not suppressed in an apoptosis-deficient host mutant. These results suggest the possibility that the apoptosis-dependent epidermal cell death and the apoptosis-independent neural malformation may represent different mechanisms underlying the Spiroplasma-induced male-killing. Despite the male-specific progressive embryonic abnormality, Spiroplasma titers remained almost constant throughout the observed stages of embryonic development and across male and female embryos. Strikingly, a few Spiroplasma-infected embryos exhibited gynandromorphism, wherein apoptotic cell death was restricted to male cells. These observations suggest that neither quantity nor proliferation of Spiroplasma cells but some Spiroplasma-derived factor(s) may be responsible for the expression of the male-killing phenotype. Symbiotic bacteria are ubiquitously associated with diverse insects, and affect their host biology in a variety of ways. In Drosophila fruit flies, infection with Spiroplasma symbionts often causes male-specific embryonic mortality, resulting in the production of all-female offspring. This striking phenotype is called “male-killing”, whose underlying mechanisms are of great interest. Here we investigated Drosophila melanogaster and its native Spiroplasma symbiont strain to understand how the host's molecular, cellular and morphogenetic pathways are involved in the symbiont-induced male-killing. Specifically in Spiroplasma-infected male embryos, pathogenic phenotypes including massive cell death throughout the body and neural malformation were observed. We unequivocally identified that the male-specific cell death preferentially occurs in the embryonic epithelium via the host's apoptotic pathway. Meanwhile, we found that, unexpectedly, the male-specific neural defects occur independently of host's apoptosis, suggesting that at least two different mechanisms may be involved in the Spiroplasma-induced male-killing. Also unexpected was the finding that Spiroplasma titers are almost constant throughout embryogenesis irrespective of sex despite the male-specific severe apoptosis. We serendipitously found Spiroplasma-infected sexual mosaic embryos, wherein apoptosis was associated with male cells, which suggests that some Spiroplasma-derived factor(s) may selectively act on male cells and cause male-killing.
Collapse
|
8
|
|
9
|
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]
|
10
|
Abstract
The caspases, a family of cysteine proteases, function as central regulators of cell death. Recently, caspase activity and caspase substrates identified in the absence of cell death have sparked strong interest in caspase functions in nonapoptotic cellular responses; these functions suggest that caspases may be activated without inducing or before apoptosis, thus leading to the cleavage of a specific subset of substrates. This review focuses primarily on the caspase enzymatic activity. Detailed genetic analyses of caspase-deficient Caenorhabditis elegans, Drosophila, and mice have shown that caspases are essential, not only for controlling the number of cells involved in sculpting or deleting structures in developing animals, but also for dynamic, nonapoptotic cell processes, such as innate immune response, tissue regeneration, cell-fate determination, stem-cell differentiation and neural activation. Our understanding of the spatio-temporal caspase activation mechanisms has advanced, primarily through the study of Drosophila developmental processes. This review will discuss current findings regarding caspase functions in cytoskeletal modification, morphogenetic regulation of cell shape, cell migration and the production of mechanical force during embryogenesis.
Collapse
Affiliation(s)
- Erina Kuranaga
- Laboratory for Histogenetic Dynamics, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
11
|
D'Amelio M, Sheng M, Cecconi F. Caspase-3 in the central nervous system: beyond apoptosis. Trends Neurosci 2012; 35:700-9. [PMID: 22796265 DOI: 10.1016/j.tins.2012.06.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 06/14/2012] [Accepted: 06/15/2012] [Indexed: 01/06/2023]
Abstract
Caspase-3 has been identified as a key mediator of neuronal programmed cell death. This protease plays a central role in the developing nervous system and its activation is observed early in neural tube formation and persists during postnatal differentiation of the neural network. Caspase-3 activation, a crucial event of neuronal cell death program, is also a feature of many chronic neurodegenerative diseases. This traditional apoptotic function of caspase-3 is challenged by recent studies that reveal new cell death-independent roles for mitochondrial-activated caspase-3 in neurite pruning and synaptic plasticity. These findings underscore the need for further research into the mechanism of action and functions of caspase-3 that may prove useful in the development of novel pharmacological treatments for a diverse range of neurological disorders.
Collapse
Affiliation(s)
- Marcello D'Amelio
- Istituto di Ricovero e Cura a Carattere Scientifico, S. Lucia Foundation, via del Fosso di Fiorano 65, 00143 Rome, Italy.
| | | | | |
Collapse
|
12
|
Miura M. Apoptotic and nonapoptotic caspase functions in animal development. Cold Spring Harb Perspect Biol 2012; 4:4/10/a008664. [PMID: 23028118 DOI: 10.1101/cshperspect.a008664] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A developing animal is exposed to both intrinsic and extrinsic stresses. One stress response is caspase activation. Caspase activation not only controls apoptosis but also proliferation, differentiation, cell shape, and cell migration. Caspase activation drives development by executing cell death or nonapoptotic functions in a cell-autonomous manner, and by secreting signaling molecules or generating mechanical forces, in a noncell autonomous manner.
Collapse
Affiliation(s)
- Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, and CREST, JST, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
13
|
Hippo signalling controls Dronc activity to regulate organ size in Drosophila. Cell Death Differ 2012; 19:1664-76. [PMID: 22555454 DOI: 10.1038/cdd.2012.48] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Hippo pathway controls organ size by multiple mechanisms that ultimately regulate the transcriptional co-activator Yorkie (Yki). Downregulation of Hippo signalling leads to tissue overgrowths due to Yki-mediated activation of target genes, whereas overexpression of the pathway triggers apoptosis in developing tissues. However, the mechanism underlying cell death induced by Hippo (Hpo)-activation is not understood. We found that overexpression of Hpo leads to induction of Dronc (Drosophila Caspase-9 homologue) expression and downregulation of dronc can suppress/block Hpo-mediated apoptosis. Furthermore, upregulation of Dronc activity strongly suppressed the overgrowth caused by Yki overexpression thereby suggesting that Hippo signalling restricts Dronc activity. Hippo-mediated cell death requires the activity of the initiator caspase Dronc. Loss-of-function of dronc in genetic mosaics leads to cell survival and increased cell proliferation in imaginal discs. dronc is transcriptionally suppressed in Yki overexpressing cells or cells mutant for other Hippo pathway components like warts (wts). We propose that Dronc is a transcriptional target of the Hippo signalling pathway. The Hippo-Dronc connection has implications in control of overall organ size and other growth regulatory mechanisms like compensatory proliferation and cell competition.
Collapse
|
14
|
Florentin A, Arama E. Caspase levels and execution efficiencies determine the apoptotic potential of the cell. ACTA ACUST UNITED AC 2012; 196:513-27. [PMID: 22351928 PMCID: PMC3283987 DOI: 10.1083/jcb.201107133] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Differences in expression level of the effector caspases Drice and Dcp-1 and in their intrinsic abilities to induce apoptosis and to control the rate of cell death underlie the differential sensitivities of cells to apoptosis. Essentially, all metazoan cells can undergo apoptosis, but some cells are more sensitive than others to apoptotic stimuli. To date, it is unclear what determines the apoptotic potential of the cell. We set up an in vivo system for monitoring and comparing the activity levels of the two main effector caspases in Drosophila melanogaster, Drice and Dcp-1. Both caspases were activated by the apoptosome after irradiation. However, whereas each caspase alone could induce apoptosis, Drice was a more effective inducer of apoptosis than Dcp-1, which instead had a role in establishing the rate of cell death. These functional differences are attributed to their intrinsic properties rather than merely their tissue specificities. Significantly, the levels of the procaspases are directly proportional to their activity levels and play a key role in determining the cell’s sensitivity to apoptosis. Finally, we provide evidence for the existence of a cellular execution threshold of caspase activity, which must be reached to induce apoptosis.
Collapse
Affiliation(s)
- Anat Florentin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
15
|
Tian L, Liu S, Liu H, Li S. 20-hydroxyecdysone upregulates apoptotic genes and induces apoptosis in the Bombyx fat body. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2012; 79:207-219. [PMID: 22517444 DOI: 10.1002/arch.20457] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
During insect metamorphosis, obsolete larval tissues are removed by programed cell death (PCD), mainly apoptosis and autophagy, which is directed by the molting hormone, 20-hydroxyecdysone (20E) and the 20E-triggered transcriptional cascade. Here, we investigated how 20E regulates apoptosis at the transcriptional level in the fat body of the silkworm, Bombyx mori. As detected by TdT-mediated dUTP Nick-End Labeling (TUNEL), apoptosis weakly occurred during the fourth larval molting, decreased to undetected levels during the early fifth instar, and gradually increased from day 4 of fifth instar to the wandering stage to the prepupal stage. Meanwhile, as determined by quantitative real-time PCR, eight genes involved in apoptosis, including Apaf-1, Nedd2 like1, Nedd2 like2, ICE1, ICE3, ICE5, Arp, and IAP, were highly expressed during molting and pupation, when the 20E titer is high. Injection of 20E into day 2 of fifth instar larvae significantly induced apoptosis and upregulated apoptotic genes after 6 h of treatment, and in vitro treatment of larval fat body tissues with 20E upregulated all the eight apoptotic genes. Moreover, RNAi knockdown of USP, a component of the 20E receptor complex EcR-USP, at the early-wandering stage reduced apoptosis and downregulated apoptotic genes after 24 h of treatment. Taken together, we infer that 20E upregulates apoptotic genes and thus induces apoptosis in the Bombyx fat body during larval molting and the larval-pupal transition.
Collapse
Affiliation(s)
- Ling Tian
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | | | | | | |
Collapse
|
16
|
Effects of manipulating apoptosis on Sindbis virus infection of Aedes aegypti mosquitoes. J Virol 2012; 86:6546-54. [PMID: 22438551 DOI: 10.1128/jvi.00125-12] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Improved control of vector-borne diseases requires an understanding of the molecular factors that determine vector competence. Apoptosis has been shown to play a role in defense against viruses in insects and mammals. Although some observations suggest a correlation between apoptosis and resistance to arboviruses in mosquitoes, there is no direct evidence tying apoptosis to arbovirus vector competence. To determine whether apoptosis can influence arbovirus replication in mosquitoes, we manipulated apoptosis in Aedes aegypti mosquitoes by silencing the expression of genes that either positively or negatively regulate apoptosis. Silencing of the A. aegypti anti-apoptotic gene iap1 (Aeiap1) caused apoptosis in midgut epithelium, alterations in midgut morphology, and 60 to 70% mosquito mortality. Mortality induced by Aeiap1 silencing was rescued by cosilencing the initiator caspase gene Aedronc, indicating that the mortality was due to apoptosis. When mosquitoes which had been injected with Aeiap1 double-stranded RNA (dsRNA) were orally infected with Sindbis virus (SINV), increased midgut infection and virus dissemination to other organs were observed. This increase in virus infection may have been due to the effects of widespread apoptosis on infection barriers or innate immunity. In contrast, silencing the expression of Aedronc, which would be expected to inhibit apoptosis, reduced SINV midgut infection and virus dissemination. Thus, our data suggest that some level of caspase activity and/or apoptosis may be necessary for efficient virus replication and dissemination in mosquitoes. This is the first study to directly test the roles of apoptosis and caspases in determining mosquito vector competence for arboviruses.
Collapse
|
17
|
Kaneko Y, Yasanga T, Suzuki M, Sakurai S. Larval fat body cells die during the early pupal stage in the frame of metamorphosis remodelation in Bombyx mori. JOURNAL OF INSECT PHYSIOLOGY 2011; 57:1715-1722. [PMID: 21971017 DOI: 10.1016/j.jinsphys.2011.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/15/2011] [Accepted: 09/22/2011] [Indexed: 05/31/2023]
Abstract
In holometabolus insects, morphology of the larval fat body is remodeled during metamorphosis. In higher Diptera, remodeling of the fat body is achieved by cell death of larval fat body cells and differentiation of the adult fat body from primordial cells. However, little is known about remodeling of the fat body at pupal metamorphosis in Lepidoptera. In this study, we found that cell death of the larval fat body in Bombyx mori occurs at shortly after pupation. About 30% of the fat body cells underwent cell death on days 1 and 2 after pupation. The cell death involved genomic DNA fragmentation, a characteristic of apoptosis. Surgical manipulation and in vitro culture of fat body cells revealed that 20-hydroxyecdysone and juvenile hormone had no effect on either initiation or progression of cell death. During cell death, a large increase in activity of caspase-3, a key enzyme of cell death, was observed. Western blot analysis of the active form of caspase-3-like protein revealed that the length of caspase-3 of B. mori was much larger than that of caspase-3 in other species. The results suggest that larval fat body cells of B. mori are removed through cell death, which is mediated by a caspase probably categorized in a novel family.
Collapse
Affiliation(s)
- Yu Kaneko
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Kakumamachi, Kanazawa 920-1192, Japan.
| | | | | | | |
Collapse
|
18
|
Drosophila IAP1-mediated ubiquitylation controls activation of the initiator caspase DRONC independent of protein degradation. PLoS Genet 2011; 7:e1002261. [PMID: 21909282 PMCID: PMC3164697 DOI: 10.1371/journal.pgen.1002261] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/06/2011] [Indexed: 02/07/2023] Open
Abstract
Ubiquitylation targets proteins for proteasome-mediated degradation and plays important roles in many biological processes including apoptosis. However, non-proteolytic functions of ubiquitylation are also known. In Drosophila, the inhibitor of apoptosis protein 1 (DIAP1) is known to ubiquitylate the initiator caspase DRONC in vitro. Because DRONC protein accumulates in diap1 mutant cells that are kept alive by caspase inhibition (“undead” cells), it is thought that DIAP1-mediated ubiquitylation causes proteasomal degradation of DRONC, protecting cells from apoptosis. However, contrary to this model, we show here that DIAP1-mediated ubiquitylation does not trigger proteasomal degradation of full-length DRONC, but serves a non-proteolytic function. Our data suggest that DIAP1-mediated ubiquitylation blocks processing and activation of DRONC. Interestingly, while full-length DRONC is not subject to DIAP1-induced degradation, once it is processed and activated it has reduced protein stability. Finally, we show that DRONC protein accumulates in “undead” cells due to increased transcription of dronc in these cells. These data refine current models of caspase regulation by IAPs. The Drosophila inhibitor of apoptosis 1 (DIAP1) readily promotes ubiquitylation of the CASPASE-9–like initiator caspase DRONC in vitro and in vivo. Because DRONC protein accumulates in diap1 mutant cells that are kept alive by effector caspase inhibition—producing so-called “undead” cells—it has been proposed that DIAP1-mediated ubiquitylation would target full-length DRONC for proteasomal degradation, ensuring survival of normal cells. However, this has never been tested rigorously in vivo. By examining loss and gain of diap1 function, we show that DIAP1-mediated ubiquitylation does not trigger degradation of full-length DRONC. Our analysis demonstrates that DIAP1-mediated ubiquitylation controls DRONC processing and activation in a non-proteolytic manner. Interestingly, once DRONC is processed and activated, it has reduced protein stability. We also demonstrate that “undead” cells induce transcription of dronc, explaining increased protein levels of DRONC in these cells. This study re-defines the mechanism by which IAP-mediated ubiquitylation regulates caspase activity.
Collapse
|
19
|
Abstract
The caspases are a family of cysteine proteases that function as central regulators of cell death. Recent investigations in Caenorhabditis elegans, Drosophila, and mice indicate that caspases are essential not only in controlling the number of cells involved in sculpting or deleting structures in developing animals, but also in dynamic cell processes such as cell-fate determination, compensatory proliferation of neighboring cells, and actin cytoskeleton reorganization, in a non-apoptotic context during development. This review focuses primarily on caspase functions involving their enzymatic activity.
Collapse
Affiliation(s)
- Erina Kuranaga
- Laboratory for Histogenetic Dynamics, RIKEN Center for Developmental Biology, Kobe, 650-0047, Japan.
| |
Collapse
|
20
|
Ouyang Y, Petritsch C, Wen H, Jan L, Jan YN, Lu B. Dronc caspase exerts a non-apoptotic function to restrain phospho-Numb-induced ectopic neuroblast formation in Drosophila. Development 2011; 138:2185-96. [PMID: 21558368 DOI: 10.1242/dev.058347] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Drosophila neuroblasts have served as a model to understand how the balance of stem cell self-renewal versus differentiation is achieved. Drosophila Numb protein regulates this process through its preferential segregation into the differentiating daughter cell. How Numb restricts the proliferation and self-renewal potentials of the recipient cell remains enigmatic. Here, we show that phosphorylation at conserved sites regulates the tumor suppressor activity of Numb. Enforced expression of a phospho-mimetic form of Numb (Numb-TS4D) or genetic manipulation that boosts phospho-Numb levels, attenuates endogenous Numb activity and causes ectopic neuroblast formation (ENF). This effect on neuroblast homeostasis occurs only in the type II neuroblast lineage. We identify Dronc caspase as a novel binding partner of Numb, and demonstrate that overexpression of Dronc suppresses the effects of Numb-TS4D in a non-apoptotic and possibly non-catalytic manner. Reduction of Dronc activity facilitates ENF induced by phospho-Numb. Our findings uncover a molecular mechanism that regulates Numb activity and suggest a novel role for Dronc caspase in regulating neural stem cell homeostasis.
Collapse
Affiliation(s)
- Yingshi Ouyang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
21
|
Winbush A, Weeks JC. Steroid-triggered, cell-autonomous death of a Drosophila motoneuron during metamorphosis. Neural Dev 2011; 6:15. [PMID: 21521537 PMCID: PMC3098771 DOI: 10.1186/1749-8104-6-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 04/27/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The metamorphosis of Drosophila melanogaster is accompanied by elimination of obsolete neurons via programmed cell death (PCD). Metamorphosis is regulated by ecdysteroids, including 20-hydroxyecdysone (20E), but the roles and modes of action of hormones in regulating neuronal PCD are incompletely understood. RESULTS We used targeted expression of GFP to track the fate of a larval motoneuron, RP2, in ventral ganglia. RP2s in abdominal neuromeres two through seven (A2 to A7) exhibited fragmented DNA by 15 hours after puparium formation (h-APF) and were missing by 20 h-APF. RP2 death began shortly after the 'prepupal pulse' of ecdysteroids, during which time RP2s expressed ecdysteroid receptors (EcRs). Genetic manipulations showed that RP2 death required the function of EcR-B isoforms, the death-activating gene, reaper (but not hid), and the apoptosome component, Dark. PCD was blocked by expression of the caspase inhibitor p35 but unaffected by manipulating Diap1. In contrast, aCC motoneurons in neuromeres A2 to A7, and RP2s in neuromere A1, expressed EcRs during the prepupal pulse but survived into the pupal stage under all conditions tested. To test the hypothesis that ecdysteroids trigger RP2's death directly, we placed abdominal GFP-expressing neurons in cell culture immediately prior to the prepupal pulse, with or without 20E. 20E induced significant PCD in putative RP2s, but not in control neurons, as assessed by morphological criteria and propidium iodide staining. CONCLUSIONS These findings suggest that the rise of ecdysteroids during the prepupal pulse acts directly, via EcR-B isoforms, to activate PCD in RP2 motoneurons in abdominal neuromeres A2 to A7, while sparing RP2s in A1. Genetic manipulations suggest that RP2's death requires Reaper function, apoptosome assembly and Diap1-independent caspase activation. RP2s offer a valuable 'single cell' approach to the molecular understanding of neuronal death during insect metamorphosis and, potentially, of neurodegeneration in other contexts.
Collapse
Affiliation(s)
- Ari Winbush
- Department of Biology, Institute of Neuroscience, University of Oregon Eugene, OR, 97403-1254, USA
| | | |
Collapse
|
22
|
Lee G, Wang Z, Sehgal R, Chen CH, Kikuno K, Hay B, Park JH. Drosophila caspases involved in developmentally regulated programmed cell death of peptidergic neurons during early metamorphosis. J Comp Neurol 2011; 519:34-48. [PMID: 21120926 DOI: 10.1002/cne.22498] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A great number of obsolete larval neurons in the Drosophila central nervous system are eliminated by developmentally programmed cell death (PCD) during early metamorphosis. To elucidate the mechanisms of neuronal PCD occurring during this period, we undertook genetic dissection of seven currently known Drosophila caspases in the PCD of a group of interneurons (vCrz) that produce corazonin (Crz) neuropeptide in the ventral nerve cord. The molecular death program in the vCrz neurons initiates within 1 hour after pupariation, as demonstrated by the cytological signs of cell death and caspase activation. PCD was significantly suppressed in dronc-null mutants, but not in null mutants of either dredd or strica. A double mutation lacking both dronc and strica impaired PCD phenotype more severely than did a dronc mutation alone, but comparably to a triple dredd/strica/dronc mutation, indicating that dronc is a main initiator caspase, while strica plays a minor role that overlaps with dronc's. As for effector caspases, vCrz PCD requires both ice and dcp-1 functions, as they work cooperatively for a timely removal of the vCrz neurons. Interestingly, the activation of the Ice and Dcp-1 is not solely dependent on Dronc and Strica, implying an alternative pathway to activate the effectors. Two remaining effector caspase genes, decay and damm, found no apparent functions in the neuronal PCD, at least during early metamorphosis. Overall, our work revealed that vCrz PCD utilizes dronc, strica, dcp-1, and ice wherein the activation of Ice and Dcp-1 requires a novel pathway in addition to the initiator caspases.
Collapse
Affiliation(s)
- Gyunghee Lee
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Apoptotic and Non-apoptotic Caspase Functions in Neural Development. Neurochem Res 2010; 36:1253-60. [DOI: 10.1007/s11064-010-0341-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2010] [Indexed: 02/08/2023]
|
24
|
Metabolic regulation of Drosophila apoptosis through inhibitory phosphorylation of Dronc. EMBO J 2010; 29:3196-207. [PMID: 20700104 DOI: 10.1038/emboj.2010.191] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 07/15/2010] [Indexed: 11/09/2022] Open
Abstract
Apoptosis ensures tissue homeostasis in response to developmental cues or cellular damage. Recently reported genome-wide RNAi screens have suggested that several metabolic regulators can modulate caspase activation in Drosophila. Here, we establish a previously unrecognized link between metabolism and Drosophila apoptosis by showing that cellular NADPH levels modulate the initiator caspase Dronc through its phosphorylation at S130. Depletion of NADPH removed this inhibitory phosphorylation, resulting in the activation of Dronc and subsequent cell death. Conversely, upregulation of NADPH prevented Dronc-mediated apoptosis upon DIAP1 RNAi or cycloheximide treatment. Furthermore, this CaMKII-mediated phosphorylation of Dronc hindered Dronc activation, but not its catalytic activity. Blockade of NADPH production aggravated the death-inducing activity of Dronc in specific neurons, but not in the photoreceptor cells of the eyes of transgenic flies; similarly, non-phosphorylatable Dronc was more potent than wild type in triggering specific neuronal apoptosis. Our observations reveal a novel regulatory circuitry in Drosophila apoptosis, and, as NADPH levels are elevated in cancer cells, also provide a genetic model to understand aberrations in cancer cell apoptosis resulting from metabolic alterations.
Collapse
|
25
|
Abstract
DNA damage or unprotected telomeres can trigger apoptosis via signaling pathways that directly sense abnormal DNA structures and activate the p53 transcription factor. We describe a p53-independent mechanism that acts in parallel to the canonical DNA damage response pathway in Drosophila to induce apoptosis after exposure to ionizing radiation. Following recovery from damage-induced cell cycle arrest, p53 mutant cells activate the JNK pathway and expression of the pro-apoptotic gene hid. Mutations in grp, a cell cycle checkpoint gene, and puc, a negative regulator of the JNK pathway, sensitize p53 mutant cells to ionizing radiation (IR)-induced apoptosis. Induction of chromosome aberrations by DNA damage generates cells with segmental aneuploidy and heterozygous for mutations in ribosomal protein genes. p53-independent apoptosis limits the formation of these aneuploid cells following DNA damage. We propose that reduced copy number of haploinsufficient genes following chromosome damage activates apoptosis and helps maintain genomic integrity.
Collapse
|
26
|
|
27
|
Xu D, Woodfield SE, Lee TV, Fan Y, Antonio C, Bergmann A. Genetic control of programmed cell death (apoptosis) in Drosophila. Fly (Austin) 2009; 3:78-90. [PMID: 19182545 PMCID: PMC2702463 DOI: 10.4161/fly.3.1.7800] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Programmed cell death, or apoptosis, is a highly conserved cellular process that has been intensively investigated in nematodes, flies and mammals. The genetic conservation, the low redundancy, the feasibility for high-throughput genetic screens and the identification of temporally and spatially regulated apoptotic responses make Drosophila melanogaster a great model for the study of apoptosis. Here, we review the key players of the cell death pathway in Drosophila and discuss their roles in apoptotic and non-apoptotic processes.
Collapse
Affiliation(s)
- Dongbin Xu
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| | - Sarah E. Woodfield
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| | - Tom V. Lee
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| | - Yun Fan
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| | - Christian Antonio
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| | - Andreas Bergmann
- The University of Texas MD Anderson Cancer Center; The Genes and Development Graduate Program; Department of Biochemistry and Molecular Biology; Houston, Texas USA; Baylor College of Medicine; Graduate Program of Developmental Biology; Houston, Texas USA
| |
Collapse
|
28
|
Parthasarathy R, Palli SR. Proliferation and differentiation of intestinal stem cells during metamorphosis of the red flour beetle,Tribolium castaneum. Dev Dyn 2008; 237:893-908. [DOI: 10.1002/dvdy.21475] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
29
|
Dorstyn L, Kumar S. A biochemical analysis of the activation of the Drosophila caspase DRONC. Cell Death Differ 2007; 15:461-70. [DOI: 10.1038/sj.cdd.4402288] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
30
|
Chitnis NS, D'Costa SM, Paul ER, Bilimoria SL. Modulation of iridovirus-induced apoptosis by endocytosis, early expression, JNK, and apical caspase. Virology 2007; 370:333-42. [PMID: 17942133 PMCID: PMC7103334 DOI: 10.1016/j.virol.2007.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Revised: 09/04/2007] [Accepted: 09/10/2007] [Indexed: 01/17/2023]
Abstract
Chilo iridescent virus (CIV) is the type species for the family Iridoviridae, which are large, isometric, cytoplasmic dsDNA viruses. We examined the mechanism of apoptosis induction by CIV. High CIV doses (CIVXS; 400 μg/ml), UV-irradiated virus (CIVUV; 10 μg/ml) and CVPE (CIV protein extract; 10 μg/ml) induced apoptosis in 60% of treated Choristoneura fumiferana (IPRI-CF-124T) cells. Normal doses of infectious CIV (10 μg/ml) induced apoptosis in only 10% of C. fumiferana (CF) cells. Apoptosis was inhibited by Z-IETD-FMK, an apical caspase inhibitor, indicating that CIV-induced apoptosis requires caspase activity. The putative caspase in CF cells was designated Cf-caspase-i. CIVUV or CVPE enhanced Cf-caspase-i activity by 80% at 24 h relative to mock-treated cells. Since the MAP kinase pathway induces or inhibits apoptosis depending on the context, we used JNK inhibitor SP600125 and demonstrated drastic suppression of CVPE-induced apoptosis. Thus, the JNK signaling pathway is significant for apoptosis in this system. Virus interaction with the cell surface was not sufficient for apoptosis since CIVUV particles bound to polysterene beads failed to induce apoptosis. Endocytosis inhibitors (bafilomycin or ammonium chloride) negated apoptosis induction by CIVUV, CIVXS or CVPE indicating that entry through this mode is required. Given the weak apoptotic response to infectious CIV, we postulated that viral gene expression inhibited apoptosis. CIV infection of cells pretreated with cycloheximide induced apoptosis in 69% of the cells compared to 10% in normal infections. Furthermore, blocking viral DNA replication with aphidicolin or phosphonoacetic acid suppressed apoptosis and Cf-caspase-i activity, indicating that early viral expression is necessary for inhibition of apoptosis, and de novo synthesis of viral proteins is not required for induction. We show for the first time that, in a member of the family Iridoviridae, apoptosis: (i) requires entry and endocytosis of virions or virion proteins, (ii) is inhibited under conditions permitting early viral expression, and (iii) requires the JNK signaling pathway. This is the first report of JNK signal requirement during apoptosis induction by an insect virus.
Collapse
Affiliation(s)
- Nilesh S Chitnis
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409-3131, USA
| | | | | | | |
Collapse
|
31
|
Cooper DM, Thi EP, Chamberlain CM, Pio F, Lowenberger C. Aedes Dronc: a novel ecdysone-inducible caspase in the yellow fever mosquito, Aedes aegypti. INSECT MOLECULAR BIOLOGY 2007; 16:563-72. [PMID: 17725799 DOI: 10.1111/j.1365-2583.2007.00758.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Caspases are cysteinyl-aspartate-specific proteases known for their role in apoptosis. Here, we describe the characterization of Aedes Dronc, a novel caspase in the yellow fever mosquito, Aedes aegypti. Aedes Dronc is predicted to contain an N-terminal caspase recruitment domain and is a homologue of Drosophila Dronc and human caspase-9. An increase in transcripts and caspase activity coincides with developmental changes in the mosquito, suggesting that Aedes Dronc plays a role in developmental apoptosis. Exposure of third instar larvae to ecdysone resulted in a significant increase in both transcript levels and caspase activity. We present here a functional characterization of the first caspase recruitment domain-containing caspase in mosquitoes, and will initiate studies on the role of apoptosis in the innate immune response of vectors.
Collapse
Affiliation(s)
- D M Cooper
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada.
| | | | | | | | | |
Collapse
|
32
|
Baum JS, Arama E, Steller H, McCall K. The Drosophila caspases Strica and Dronc function redundantly in programmed cell death during oogenesis. Cell Death Differ 2007; 14:1508-17. [PMID: 17464325 DOI: 10.1038/sj.cdd.4402155] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Programmed cell death (PCD) in the Drosophila ovary occurs either during mid-oogenesis, resulting in degeneration of the entire egg chamber or during late oogenesis, to facilitate the development of the oocyte. PCD during oogenesis is regulated by mechanisms different from those that control cell death in other Drosophila tissues. We have analyzed the role of caspases in PCD of the female germline by examining caspase mutants and overexpressing caspase inhibitors. Imprecise P-element excision was used to generate mutants of the initiator caspase strica. While null mutants of strica or another initiator caspase, dronc, display no ovary phenotype, we find that strica exhibits redundancy with dronc, during both mid- and late oogenesis. Ovaries of double mutants contain defective mid-stage egg chambers similar to those reported previously in dcp-1 mutants, and mature egg chambers with persisting nurse cell nuclei. In addition, the effector caspases drice and dcp-1 also display redundant functions during late oogenesis, resulting in persisting nurse cell nuclei. These findings indicate that caspases are required for nurse cell death during mid-oogenesis, and participate in developmental nurse cell death during late oogenesis. This reveals a novel pathway of cell death in the ovary that utilizes strica, dronc, dcp-1 and drice, and importantly illustrates strong redundancy among the caspases.
Collapse
Affiliation(s)
- J S Baum
- Department of Biology, Graduate Program, Boston University, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
33
|
Peterson JS, Bass BP, Jue D, Rodriguez A, Abrams JM, McCall K. Noncanonical cell death pathways act during Drosophila oogenesis. Genesis 2007; 45:396-404. [PMID: 17506088 DOI: 10.1002/dvg.20306] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Programmed cell death (PCD) is a highly conserved process that occurs during development and in response to adverse conditions. In Drosophila, most PCDs require the genes within the H99 deficiency, the adaptor molecule Ark, and caspases. Here we investigate 10 cell death genes for their potential roles in two distinct types of PCD that occur in oogenesis: developmental nurse cell PCD and starvation-induced PCD. Most of the genes investigated were found to have little effect on late stage developmental PCD in oogenesis, although ark mutants showed a partial inhibition. Mid-stage starvation-induced germline PCD was found to be independent of the upstream activators and ark although it requires caspases, suggesting an apoptosome-independent mechanism of caspase activation in mid-oogenesis. These results indicate that novel pathways must control PCD in the ovary.
Collapse
Affiliation(s)
- Jeanne S Peterson
- Department of Biology, Boston University, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
34
|
Cao C, Liu Y, Lehmann M. Fork head controls the timing and tissue selectivity of steroid-induced developmental cell death. ACTA ACUST UNITED AC 2007; 176:843-52. [PMID: 17339378 PMCID: PMC2064058 DOI: 10.1083/jcb.200611155] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell death during Drosophila melanogaster metamorphosis is controlled by the steroid hormone 20-hydroxyecdysone (20E). Elements of the signaling pathway that triggers death are known, but it is not known why some tissues, and not others, die in response to a particular hormone pulse. We found that loss of the tissue-specific transcription factor Fork head (Fkh) is both required and sufficient to specify a death response to 20E in the larval salivary glands. Loss of fkh itself is a steroid-controlled event that is mediated by the 20E-induced BR-C gene, and that renders the key death regulators hid and reaper hormone responsive. These results implicate the D. melanogaster FOXA orthologue Fkh with a novel function as a competence factor for steroid-controlled cell death. They explain how a specific tissue is singled out for death, and why this tissue survives earlier hormone pulses. More generally, they suggest that cell identity factors like Fkh play a pivotal role in the normal control of developmental cell death.
Collapse
Affiliation(s)
- Chike Cao
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | | | | |
Collapse
|
35
|
Kuranaga E, Miura M. Nonapoptotic functions of caspases: caspases as regulatory molecules for immunity and cell-fate determination. Trends Cell Biol 2007; 17:135-44. [PMID: 17275304 DOI: 10.1016/j.tcb.2007.01.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/20/2006] [Accepted: 01/17/2007] [Indexed: 11/30/2022]
Abstract
Caspases are a family of cysteine proteases that are highly conserved in multicellular organisms and function as central regulators of apoptosis. Recent investigations in Caenorhabditis elegans, Drosophila and mice suggest that caspases also function as regulatory molecules for immunity and cell-fate determination. Here, we review genetic studies of nonapoptotic functions of caspases and discuss the regulatory mechanisms of caspases for executing nonapoptotic functions.
Collapse
Affiliation(s)
- Erina Kuranaga
- Department of Genetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | |
Collapse
|
36
|
Parthasarathy R, Palli SR. Developmental and hormonal regulation of midgut remodeling in a lepidopteran insect, Heliothis virescens. Mech Dev 2007; 124:23-34. [PMID: 17107775 DOI: 10.1016/j.mod.2006.09.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 09/12/2006] [Accepted: 09/16/2006] [Indexed: 11/22/2022]
Abstract
Midgut tissue undergoes remodeling during metamorphosis in insects belonging to orders Lepidoptera and Diptera. We investigated the developmental and hormonal regulation of these remodeling events in lepidopteran insect, Heliothis virescens. In H. virescens, programmed cell death (PCD) of larval midgut cells as well as proliferation and differentiation of imaginal cells began at 108 h after ecdysis to the final larval instar (AEFL) and proceeded through the pupal stages. Expression patterns of pro- cell death factors (caspase-1 and ICE) and anti-cell death factor, Inhibitor of Apoptosis (IAP) were studied in midguts during last larval and pupal stages. IAP, Caspase-1 and ICE mRNAs showed peaks at 48 h AEFL, 96 h AEFL and in newly formed pupae, respectively. Immunohistochemical analysis substantiated high caspase-3 activity in midgut at 108 h AEFL. Application of methoprene, a juvenile hormone analog (JHA) blocked PCD by maintaining high levels of IAP, downregulating the expression of caspase-1, ICE and inhibiting an increase in caspase-3 protein levels in midgut tissue. Also, the differentiation of imaginal cells was impaired by methoprene treatment. These studies demonstrate that presence of JHA during final instar larvae affects both midgut remodeling and larval-pupal metamorphosis leading to larval/pupal deformities in lepidopteran insects, a mechanism that is different from that in mosquito, Ae. aegypti where JHA uncouples midgut remodeling from metamorphosis.
Collapse
Affiliation(s)
- R Parthasarathy
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, KY 40546, USA
| | | |
Collapse
|
37
|
Abstract
Cell death plays many roles during development, in the adult, and in the genesis of many pathological states. Much of this death is apoptotic in nature and requires the activity of members of the caspase family of proteases. It is now possible uniquely in Drosophila to carry out genetic screens for genes that determine the fate-life or death-of any population of cells during development and adulthood. This, in conjunction with the ability to obtain biochemical quantities of material, has made Drosophila a useful organism for exploring the mechanisms by which apoptosis is carried out and regulated. This review summarizes our knowledge of caspase-dependent cell death in Drosophila and compares that knowledge with what is known in worms and mammals. We also discuss the significance of recent work showing that a number of key cell death activators also play nonapoptotic roles. We highlight opportunities and outstanding questions along the way.
Collapse
Affiliation(s)
- Bruce A Hay
- Division of Biology, California Institute of Technology, Pasadena, California 91125, USA.
| | | |
Collapse
|
38
|
Abstract
The first proapoptotic caspase, CED-3, was cloned from Caenorhabditis elegans in 1993 and shown to be essential for the developmental death of all somatic cells. Following the discovery of CED-3, caspases have been cloned from several vertebrate and invertebrate species. As reviewed in other articles in this issue of Cell Death and Differentiation, many caspases function in nonapoptotic pathways. However, as is clear from the worm studies, the evolutionarily conserved role of caspases is to execute programmed cell death. In this article, I will specifically focus on caspases that function primarily in cell death execution. In particular, the physiological function of caspases in apoptosis is discussed using examples from the worm, fly and mammals.
Collapse
Affiliation(s)
- S Kumar
- Hanson Institute, Institute of Medical and Veterinary Science, Adelaide, SA, Australia.
| |
Collapse
|
39
|
Kondo S, Senoo-Matsuda N, Hiromi Y, Miura M. DRONC coordinates cell death and compensatory proliferation. Mol Cell Biol 2006; 26:7258-68. [PMID: 16980627 PMCID: PMC1592896 DOI: 10.1128/mcb.00183-06] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accidental cell death often leads to compensatory proliferation. In Drosophila imaginal discs, for example, gamma-irradiation induces extensive cell death, which is rapidly compensated by elevated proliferation. Excessive compensatory proliferation can be artificially induced by "undead cells" that are kept alive by inhibition of effector caspases in the presence of apoptotic stimuli. This suggests that compensatory proliferation is induced by dying cells as part of the apoptosis program. Here, we provide genetic evidence that the Drosophila initiator caspase DRONC governs both apoptosis execution and subsequent compensatory proliferation. We examined mutants of five Drosophila caspases and identified the initiator caspase DRONC and the effector caspase DRICE as crucial executioners of apoptosis. Artificial compensatory proliferation induced by coexpression of Reaper and p35 was completely suppressed in dronc mutants. Moreover, compensatory proliferation after gamma-irradiation was enhanced in drice mutants, in which DRONC is activated but the cells remain alive. These results show that the apoptotic pathway bifurcates at DRONC and that DRONC coordinates the execution of cell death and compensatory proliferation.
Collapse
Affiliation(s)
- Shu Kondo
- Department of Genetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | |
Collapse
|
40
|
Leulier F, Ribeiro PS, Palmer E, Tenev T, Takahashi K, Robertson D, Zachariou A, Pichaud F, Ueda R, Meier P. Systematic in vivo RNAi analysis of putative components of the Drosophila cell death machinery. Cell Death Differ 2006; 13:1663-74. [PMID: 16485033 DOI: 10.1038/sj.cdd.4401868] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Despite the identification of numerous key players of the cell death machinery, little is known about their physiological role. Using RNA interference (RNAi) in vivo, we have studied the requirement of all Drosophila caspases and caspase-adaptors in different paradigms of apoptosis. Of the seven caspases, Dronc, drICE, Strica and Decay are rate limiting for apoptosis. Surprisingly, Hid-mediated apoptosis requires a broader range of caspases than apoptosis initiated by loss of the caspase inhibitor DIAP1, suggesting that Hid causes apoptosis not only by antagonizing DIAP1 but also by activating DIAP1-independent caspase cascades. While Hid killing requires Strica, Decay, Dronc/Dark and drICE, apoptosis triggered by DIAP1 depletion merely relied upon Dronc/Dark and drICE. Furthermore, we found that overexpression of DIAP2 can rescue diap1-RNAi-mediated apoptosis, suggesting that DIAP2 regulates caspases directly. Consistently, we show that DIAP2 binds active drICE. Since DIAP2 associates with Hid, we propose a model whereby Hid co-ordinately targets both DIAP1 and DIAP2 to unleash drICE.
Collapse
Affiliation(s)
- F Leulier
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Xu D, Wang Y, Willecke R, Chen Z, Ding T, Bergmann A. The effector caspases drICE and dcp-1 have partially overlapping functions in the apoptotic pathway in Drosophila. Cell Death Differ 2006; 13:1697-706. [PMID: 16645642 PMCID: PMC2519037 DOI: 10.1038/sj.cdd.4401920] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Caspases are essential components of the apoptotic machinery in both vertebrates and invertebrates. Here, we report the isolation of a mutant allele of the Drosophila effector caspase drICE as a strong suppressor of hid- (head involution defective-) induced apoptosis. This mutant was used to determine the apoptotic role of drICE. Our data are consistent with an important function of drICE for developmental and irradiation-induced cell death. Epistatic analysis suggests that drICE acts genetically downstream of Drosophila inhibitor of apoptosis protein 1 (Diap1). However, although cell death is significantly reduced in drICE mutants in all assays, it is not completely blocked. A double-mutant analysis between drICE and death caspase-1 (dcp-1), another effector caspase, reveals that some cells (type I) strictly require drICE for apoptosis, whereas other cells (type II) require either drICE or dcp-1. Thus, these data demonstrate a barely appreciated complexity in the apoptotic pathway, and are consistent with current models about effector caspase regulation in both vertebrates and invertebrates.
Collapse
Affiliation(s)
- D Xu
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1000, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
42
|
Velentzas AD, Nezis IP, Stravopodis DJ, Papassideri IS, Margaritis LH. Mechanisms of programmed cell death during oogenesis in Drosophila virilis. Cell Tissue Res 2006; 327:399-414. [PMID: 17004067 DOI: 10.1007/s00441-006-0298-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2006] [Accepted: 06/29/2006] [Indexed: 12/01/2022]
Abstract
We describe the features of programmed cell death occurring in the egg chambers of Drosophila virilis during mid-oogenesis and late oogenesis. During mid-oogenesis, the spontaneously degenerating egg chambers exhibit typical characteristics of apoptotic cell death. As revealed by propidium iodide, rhodamine-conjugated phalloidin staining, and the TUNEL assay, respectively, the nurse cells contain condensed chromatin, altered actin cytoskeleton, and fragmented DNA. In vitro caspase activity assays and immunostaining procedures demonstrate that the atretic egg chambers possess high levels of caspase activity. Features of autophagic cell death are also observed during D. virilis mid-oogenesis, as shown by monodansylcadaverine staining, together with an ultrastructural examination by transmission electron microscopy. During the late stages of oogenesis in D. virilis, once again, the two mechanisms, viz., nurse cell cluster apoptosis and autophagy, operate together, manifesting features of cell death similar to those detailed above. Moreover, an altered form of cytochrome c seems to be released from the mitochondria in the nurse cells proximal to the oocyte. We propose that apoptosis and autophagy function synergistically during oogenesis in D. virilis in order to achieve a more efficient elimination of the degenerated nurse cells and abnormal egg chambers.
Collapse
Affiliation(s)
- Athanassios D Velentzas
- Faculty of Biology, Department of Cell Biology and Biophysics, University of Athens, Panepistimiopolis 15784, Athens, Greece
| | | | | | | | | |
Collapse
|
43
|
Colombani J, Polesello C, Josué F, Tapon N. Dmp53 activates the Hippo pathway to promote cell death in response to DNA damage. Curr Biol 2006; 16:1453-8. [PMID: 16860746 DOI: 10.1016/j.cub.2006.05.059] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 05/22/2006] [Accepted: 05/23/2006] [Indexed: 10/24/2022]
Abstract
Developmental and environmental signals control a precise program of growth, proliferation, and cell death. This program ensures that animals reach, but do not exceed, their typical size . Understanding how cells sense the limits of tissue size and respond accordingly by exiting the cell cycle or undergoing apoptosis has important implications for both developmental and cancer biology. The Hippo (Hpo) pathway comprises the kinases Hpo and Warts/Lats (Wts), the adaptors Salvador (Sav) and Mob1 as a tumor suppressor (Mats), the cytoskeletal proteins Expanded and Merlin, and the transcriptional cofactor Yorkie (Yki) . This pathway has been shown to restrict cell division and promote apoptosis. The caspase repressor DIAP1 appears to be a primary target of the Hpo pathway in cell-death control. Firstly, Hpo promotes DIAP1 phosphorylation, likely decreasing its stability. Secondly, Wts phosphorylates and inactivates Yki, decreasing DIAP1 transcription. Although we understand some of the events downstream of the Hpo kinase, its mode of activation remains mysterious. Here, we show that Hpo can be activated by Ionizing Radiations (IR) in a Dmp53 (Drosophila melanogaster p53)-dependent manner and that Hpo is required (though not absolutely) for the cell death response elicited by IR or Dmp53 ectopic expression.
Collapse
Affiliation(s)
- Julien Colombani
- Apoptosis and Proliferation Control Laboratory, Cancer Research UK, London Research Institute
| | | | | | | |
Collapse
|
44
|
Kuo CT, Zhu S, Younger S, Jan LY, Jan YN. Identification of E2/E3 ubiquitinating enzymes and caspase activity regulating Drosophila sensory neuron dendrite pruning. Neuron 2006; 51:283-90. [PMID: 16880123 DOI: 10.1016/j.neuron.2006.07.014] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Revised: 07/14/2006] [Accepted: 07/18/2006] [Indexed: 01/25/2023]
Abstract
Ubiquitin-proteasome system (UPS) is a multistep protein degradation machinery implicated in many diseases. In the nervous system, UPS regulates remodeling and degradation of neuronal processes and is linked to Wallerian axonal degeneration, though the ubiquitin ligases that confer substrate specificity remain unknown. Having shown previously that class IV dendritic arborization (C4da) sensory neurons in Drosophila undergo UPS-mediated dendritic pruning during metamorphosis, we conducted an E2/E3 ubiquitinating enzyme mutant screen, revealing that mutation in ubcD1, an E2 ubiquitin-conjugating enzyme, resulted in retention of C4da neuron dendrites during metamorphosis. Further, we found that UPS activation likely leads to UbcD1-mediated degradation of DIAP1, a caspase-antagonizing E3 ligase. This allows for local activation of the Dronc caspase, thereby preserving C4da neurons while severing their dendrites. Thus, in addition to uncovering E2/E3 ubiquitinating enzymes for dendrite pruning, this study provides a mechanistic link between UPS and the apoptotic machinery in regulating neuronal process remodeling.
Collapse
Affiliation(s)
- Chay T Kuo
- Howard Hughes Medical Institute, Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
45
|
Muro I, Berry DL, Huh JR, Chen CH, Huang H, Yoo SJ, Guo M, Baehrecke EH, Hay BA. The Drosophila caspase Ice is important for many apoptotic cell deaths and for spermatid individualization, a nonapoptotic process. Development 2006; 133:3305-15. [PMID: 16887831 DOI: 10.1242/dev.02495] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Caspase family proteases play important roles in the regulation of apoptotic cell death. Initiator caspases are activated in response to death stimuli, and they transduce and amplify these signals by cleaving and thereby activating effector caspases. In Drosophila, the initiator caspase Nc (previously Dronc) cleaves and activates two short-prodomain caspases, Dcp-1 and Ice (previously Drice), suggesting these as candidate effectors of Nc killing activity. dcp-1-null mutants are healthy and possess few defects in normally occurring cell death. To explore roles for Ice in cell death, we generated and characterized an Ice null mutant. Animals lacking Ice show a number of defects in cell death, including those that occur during embryonic development, as well as during formation of adult eyes, arista and wings. Ice mutants exhibit subtle defects in the destruction of larval tissues, and do not prevent destruction of salivary glands during metamorphosis. Cells from Ice animals are also markedly resistant to several stresses, including X-irradiation and inhibition of protein synthesis. Mutations in Ice also suppress cell death that is induced by expression of Rpr, Wrinkled (previously Hid) and Grim. These observations demonstrate that Ice plays an important non-redundant role as a cell death effector. Finally, we demonstrate that Ice participates in, but is not absolutely required for, the non-apoptotic process of spermatid differentiation.
Collapse
Affiliation(s)
- Israel Muro
- Division of Biology, MC 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wu Y, Parthasarathy R, Bai H, Palli SR. Mechanisms of midgut remodeling: juvenile hormone analog methoprene blocks midgut metamorphosis by modulating ecdysone action. Mech Dev 2006; 123:530-47. [PMID: 16829058 DOI: 10.1016/j.mod.2006.05.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Revised: 05/17/2006] [Accepted: 05/20/2006] [Indexed: 11/21/2022]
Abstract
In holometabolous insects such as mosquito, Aedes aegypti, midgut undergoes remodeling during metamorphosis. Insect metamorphosis is regulated by several hormones including juvenile hormone (JH) and 20-hydroxyecdysone (20E). The cellular and molecular events that occur during midgut remodeling were investigated by studying nuclear stained whole mounts and cross-sections of midguts and by monitoring the mRNA levels of genes involved in 20E action in methoprene-treated and untreated Ae. aegypti. We used JH analog, methoprene, to mimic JH action. In Ae. aegypti larvae, the programmed cell death (PCD) of larval midgut cells and the proliferation and differentiation of imaginal cells were initiated at about 36h after ecdysis to the 4th instar larval stage (AEFL) and were completed by 12h after ecdysis to the pupal stage (AEPS). In methoprene-treated larvae, the proliferation and differentiation of imaginal cells was initiated at 36h AEFL, but the PCD was initiated only after ecdysis to the pupal stage. However, the terminal events that occur for completion of PCD during pupal stage were blocked. As a result, the pupae developed from methoprene-treated larvae contained two midgut epithelial layers until they died during the pupal stage. Quantitative PCR analyses showed that methoprene affected midgut remodeling by modulating the expression of ecdysone receptor B, ultraspiracle A, broad complex, E93, ftz-f1, dronc and drice, the genes that are shown to play key roles in 20E action and PCD. Thus, JH analog, methoprene acts on Ae. aegypti by interfering with the expression of genes involved in 20E action resulting in a block in midgut remodeling and death during pupal stage.
Collapse
Affiliation(s)
- Yu Wu
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, KY 40546, USA
| | | | | | | |
Collapse
|
47
|
Mills K, Daish T, Harvey KF, Pfleger CM, Hariharan IK, Kumar S. The Drosophila melanogaster Apaf-1 homologue ARK is required for most, but not all, programmed cell death. ACTA ACUST UNITED AC 2006; 172:809-15. [PMID: 16533943 PMCID: PMC2063725 DOI: 10.1083/jcb.200512126] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Apaf-1 protein is essential for cytochrome c–mediated caspase-9 activation in the intrinsic mammalian pathway of apoptosis. Although Apaf-1 is the only known mammalian homologue of the Caenorhabditis elegans CED-4 protein, the deficiency of apaf-1 in cells or in mice results in a limited cell survival phenotype, suggesting that alternative mechanisms of caspase activation and apoptosis exist in mammals. In Drosophila melanogaster, the only Apaf-1/CED-4 homologue, ARK, is required for the activation of the caspase-9/CED-3–like caspase DRONC. Using specific mutants that are deficient for ark function, we demonstrate that ARK is essential for most programmed cell death (PCD) during D. melanogaster development, as well as for radiation-induced apoptosis. ark mutant embryos have extra cells, and tissues such as brain lobes and wing discs are enlarged. These tissues from ark mutant larvae lack detectable PCD. During metamorphosis, larval salivary gland removal was severely delayed in ark mutants. However, PCD occurred normally in the larval midgut, suggesting that ARK-independent cell death pathways also exist in D. melanogaster.
Collapse
Affiliation(s)
- Kathryn Mills
- Hanson Institute, Institute of Medical and Veterinary Science, Adelaide, Australia 5000
| | | | | | | | | | | |
Collapse
|
48
|
Arama E, Bader M, Srivastava M, Bergmann A, Steller H. The two Drosophila cytochrome C proteins can function in both respiration and caspase activation. EMBO J 2006; 25:232-43. [PMID: 16362035 PMCID: PMC1356363 DOI: 10.1038/sj.emboj.7600920] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 11/22/2005] [Indexed: 12/21/2022] Open
Abstract
Cytochrome C has two apparently separable cellular functions: respiration and caspase activation during apoptosis. While a role of the mitochondria and cytochrome C in the assembly of the apoptosome and caspase activation has been established for mammalian cells, the existence of a comparable function for cytochrome C in invertebrates remains controversial. Drosophila possesses two cytochrome c genes, cyt-c-d and cyt-c-p. We show that only cyt-c-d is required for caspase activation in an apoptosis-like process during spermatid differentiation, whereas cyt-c-p is required for respiration in the soma. However, both cytochrome C proteins can function interchangeably in respiration and caspase activation, and the difference in their genetic requirements can be attributed to differential expression in the soma and testes. Furthermore, orthologues of the apoptosome components, Ark (Apaf-1) and Dronc (caspase-9), are also required for the proper removal of bulk cytoplasm during spermatogenesis. Finally, several mutants that block caspase activation during spermatogenesis were isolated in a genetic screen, including mutants with defects in spermatid mitochondrial organization. These observations establish a role for the mitochondria in caspase activation during spermatogenesis.
Collapse
Affiliation(s)
- Eli Arama
- Strang Laboratory of Cancer Research, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Maya Bader
- Strang Laboratory of Cancer Research, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Mayank Srivastava
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andreas Bergmann
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hermann Steller
- Strang Laboratory of Cancer Research, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|