1
|
Damschroder D, Sun J, McDonald KO, Buttitta L. Cell cycle re-entry in the aging Drosophila brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609689. [PMID: 39253469 PMCID: PMC11383271 DOI: 10.1101/2024.08.26.609689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The brain is an organ comprised mostly of long-lived, quiescent cells that perform vital functions throughout an animal's life. Due to the brain's limited regenerative ability, these long-lived cells must engage unique mechanisms to cope with accumulated damage over time. We have shown that a subset of differentiated neuronal and glial cells in the fruit fly brain become polyploid during adulthood. Cell cycle re-entry in the brain has previously been associated with neurodegeneration, but there may be a more complex relationship between polyploidy and cell fitness in the brain. Here, we examine how known lifespan modifiers influence the accumulation of polyploidy in the aging fly brain. Flies aged at a low temperature, or with a low protein diet, accumulate polyploid cells in the brain more slowly than expected if this phenotype were solely regulated by lifespan mechanisms. Despite the slower accumulation of polyploid cells, animals under conditions that extend lifespan eventually reach similar levels of polyploidy in the brain as controls. Our work suggests known lifespan modifiers can influence the timing of cell cycle re-entry in the adult brain, indicating there is a flexible window of cell cycle plasticity in the aging brain.
Collapse
Affiliation(s)
| | - Jenny Sun
- University of Michigan, MCDB, Ann Arbor, MI 48109
| | | | | |
Collapse
|
2
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
3
|
Colombani J, Andersen DS. Drosophila TNF/TNFRs: At the crossroad between metabolism, immunity, and tissue homeostasis. FEBS Lett 2023; 597:2416-2432. [PMID: 37567762 DOI: 10.1002/1873-3468.14716] [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: 06/13/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023]
Abstract
Tumor necrosis factor (TNF)-α is a highly conserved proinflammatory cytokine with important functions in immunity, tissue repair, and cellular homeostasis. Due to the simplicity of the Drosophila TNF-TNF receptor (TNFR) system and a broad genetic toolbox, the fly has played a pivotal role in deciphering the mechanisms underlying TNF-mediated physiological and pathological functions. In this review, we summarize the recent advances in our understanding of how local and systemic sources of Egr/TNF contribute to its antitumor and tumor-promoting properties, and its emerging functions in adaptive growth responses, sleep regulation, and adult tissue homeostasis. The recent annotation of TNF as an adipokine and its indisputable contribution to obesity- and cancer-associated metabolic diseases have provoked a new area of research focusing on its dual function in regulating immunity and energy homeostasis. Here, we discuss the role of TNFR signaling in coupling immune and metabolic processes and how this might be relevant in the adaption of host to environmental stresses, or, in the case of obesity, promote metabolic derangements and disease.
Collapse
Affiliation(s)
- Julien Colombani
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ditte S Andersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Das R, Pandey P, Maurya B, Pradhan P, Sinha D, Mukherjee A, Mutsuddi M. Spoonbill positively regulates JNK signalling mediated apoptosis in Drosophila melanogaster. Eur J Cell Biol 2023; 102:151300. [PMID: 36858008 DOI: 10.1016/j.ejcb.2023.151300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
A-kinase anchoring protein (AKAP) comprises a family of scaffold proteins, which decides the subcellular localisation of a combination of signalling molecules. Spoonbill (Spoon) is a putative A-kinase anchoring protein in Drosophila. We have earlier reported that Spoon suppresses ribonuclear foci formed by trinucleotide repeat expanded transcripts associated with Spinocerebellar Ataxia 8 neurodegeneration in Drosophila. However, the role of Spoonbill in cellular signalling was unexplored. In this report, we have unravelled a novel function of Spoon protein in the regulation of the apoptotic pathway. The Drosophila TNFα homolog, Eiger, induces apoptosis via activation of the JNK pathway. We have shown here that Spoonbill is a positive regulator of the Eiger-induced JNK signalling. Further genetic interaction studies show that the spoon interacts with components of the JNK pathway, TGF-β activated kinase 1 (Tak1 - JNKKK), hemipterous (hep - JNKK) and basket (bsk - JNK). Interestingly, Spoonbill alone can also induce ectopic activation of the JNK pathway in a context-specific manner. To understand the molecular mechanism underlying Spoonbill-mediated modulation of the JNK pathway, the interaction between Spoon and Drosophila JNK was assessed. basket encodes the only known JNK in Drosophila. This serine/threonine-protein kinase phosphorylates Jra/Kay, which transcriptionally regulate downstream targets like Matrix metalloproteinase 1 (Mmp1), puckered (puc), and proapoptotic genes hid, reaper and grim. Interestingly, we found that Spoonbill colocalises and co-immunoprecipitates with the Basket protein in the developing photoreceptor neurons. Hence, we propose that Spoon plays a vital role in JNK-induced apoptosis. Furthermore, stress-induced JNK activation underlying Parkinson's Disease was also examined. In the Parkinson's Drosophila model of neurodegeneration, depletion of Spoonbill leads to a partial reduction of JNK pathway activation, along with improvement in adult motor activity. These observations suggest that the putative scaffold protein Spoonbill is a functional and physical interacting partner of the Drosophila JNK protein, Basket. Spoon protein is localised on the outer mitochondrial membrane (OMM), which may perhaps provide a suitable subcellular niche for activation of Drosophila Basket protein by its kinases which induce apoptosis.
Collapse
Affiliation(s)
- Rituparna Das
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Pranjali Pandey
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Bhawana Maurya
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | | | - Devanjan Sinha
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
5
|
Ren M, Yang Y, Heng KHY, Ng LY, Chong CYY, Ng YT, Gorur-Shandilya S, Lee RMQ, Lim KL, Zhang J, Koh TW. MED13 and glycolysis are conserved modifiers of α-synuclein-associated neurodegeneration. Cell Rep 2022; 41:111852. [PMID: 36543134 DOI: 10.1016/j.celrep.2022.111852] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/04/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
α-Synuclein (α-syn) is important in synucleinopathies such as Parkinson's disease (PD). While genome-wide association studies (GWASs) of synucleinopathies have identified many risk loci, the underlying genes have not been shown for most loci. Using Drosophila, we screened 3,471 mutant chromosomes for genetic modifiers of α-synuclein and identified 12 genes. Eleven modifiers have human orthologs associated with diseases, including MED13 and CDC27, which lie within PD GWAS loci. Drosophila Skd/Med13 and glycolytic enzymes are co-upregulated by α-syn-associated neurodegeneration. While elevated α-syn compromises mitochondrial function, co-expressing skd/Med13 RNAi and α-syn synergistically increase the ratio of oxidized-to-reduced glutathione. The resulting neurodegeneration can be suppressed by overexpressing a glycolytic enzyme or treatment with deferoxamine, suggesting that compensatory glycolysis is neuroprotective. In addition, the functional relationship between α-synuclein, MED13, and glycolytic enzymes is conserved between flies and mice. We propose that hypoxia-inducible factor and MED13 are part of a druggable pathway for PD.
Collapse
Affiliation(s)
- Mengda Ren
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308207, Singapore; National Neuroscience Institute, Singapore 308433, Singapore
| | - Ying Yang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang 310002, China
| | | | - Lu Yi Ng
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | | | - Yan Ting Ng
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore
| | | | - Rachel Min Qi Lee
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308207, Singapore; National Neuroscience Institute, Singapore 308433, Singapore
| | - Jing Zhang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang 310002, China; China National Health and Disease Human Brain Tissue Resource Center, Hangzhou, Zhejiang 310002, China
| | - Tong-Wey Koh
- Temasek Life Sciences Laboratory, Singapore 117604, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore.
| |
Collapse
|
6
|
Damage-responsive neuro-glial clusters coordinate the recruitment of dormant neural stem cells in Drosophila. Dev Cell 2022; 57:1661-1675.e7. [PMID: 35716661 DOI: 10.1016/j.devcel.2022.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022]
Abstract
Recruitment of stem cells is crucial for tissue repair. Although stem cell niches can provide important signals, little is known about mechanisms that coordinate the engagement of disseminated stem cells across an injured tissue. In Drosophila, adult brain lesions trigger local recruitment of scattered dormant neural stem cells suggesting a mechanism for creating a transient stem cell activation zone. Here, we find that injury triggers a coordinated response in neuro-glial clusters that promotes the spread of a neuron-derived stem cell factor via glial secretion of the lipocalin-like transporter Swim. Strikingly, swim is induced in a Hif1-α-dependent manner in response to brain hypoxia. Mammalian Swim (Lcn7) is also upregulated in glia of the mouse hippocampus upon brain injury. Our results identify a central role of neuro-glial clusters in promoting neural stem cell activation at a distance, suggesting a conserved function of the HIF1-α/Swim/Wnt module in connecting injury-sensing and regenerative outcomes.
Collapse
|
7
|
Coupe D, Bossing T. Insights into nervous system repair from the fruit fly. Neuronal Signal 2022; 6:NS20210051. [PMID: 35474685 PMCID: PMC9008705 DOI: 10.1042/ns20210051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Millions of people experience injury to the central nervous system (CNS) each year, many of whom are left permanently disabled, providing a challenging hurdle for the field of regenerative medicine. Repair of damage in the CNS occurs through a concerted effort of phagocytosis of debris, cell proliferation and differentiation to produce new neurons and glia, distal axon/dendrite degeneration, proximal axon/dendrite regeneration and axon re-enwrapment. In humans, regeneration is observed within the peripheral nervous system, while in the CNS injured axons exhibit limited ability to regenerate. This has also been described for the fruit fly Drosophila. Powerful genetic tools available in Drosophila have allowed the response to CNS insults to be probed and novel regulators with mammalian orthologs identified. The conservation of many regenerative pathways, despite considerable evolutionary separation, stresses that these signals are principal regulators and may serve as potential therapeutic targets. Here, we highlight the role of Drosophila CNS injury models in providing key insight into regenerative processes by exploring the underlying pathways that control glial and neuronal activation in response to insult, and their contribution to damage repair in the CNS.
Collapse
Affiliation(s)
- David Coupe
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| | - Torsten Bossing
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| |
Collapse
|
8
|
Dpp and Hedgehog promote the glial response to neuronal apoptosis in the developing Drosophila visual system. PLoS Biol 2021; 19:e3001367. [PMID: 34379617 PMCID: PMC8396793 DOI: 10.1371/journal.pbio.3001367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/27/2021] [Accepted: 07/16/2021] [Indexed: 11/19/2022] Open
Abstract
Damage in the nervous system induces a stereotypical response that is mediated by glial cells. Here, we use the eye disc of Drosophila melanogaster as a model to explore the mechanisms involved in promoting glial cell response after neuronal cell death induction. We demonstrate that these cells rapidly respond to neuronal apoptosis by increasing in number and undergoing morphological changes, which will ultimately grant them phagocytic abilities. We found that this glial response is controlled by the activity of Decapentaplegic (Dpp) and Hedgehog (Hh) signalling pathways. These pathways are activated after cell death induction, and their functions are necessary to induce glial cell proliferation and migration to the eye discs. The latter of these 2 processes depend on the function of the c-Jun N-terminal kinase (JNK) pathway, which is activated by Dpp signalling. We also present evidence that a similar mechanism controls glial response upon apoptosis induction in the leg discs, suggesting that our results uncover a mechanism that might be involved in controlling glial cells response to neuronal cell death in different regions of the peripheral nervous system (PNS). In reaction to neuronal damage, glial cells proliferate, change their morphology and alter their behaviour; this response is associated with glial cells’ regenerative function and is necessary to preserve the structural integrity and function of the nervous system. This study identifies a role for the Decapentaplegic and Hedgehog pathways in controlling the glial response.
Collapse
|
9
|
Li G, Hidalgo A. The Toll Route to Structural Brain Plasticity. Front Physiol 2021; 12:679766. [PMID: 34290618 PMCID: PMC8287419 DOI: 10.3389/fphys.2021.679766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/02/2021] [Indexed: 11/13/2022] Open
Abstract
The human brain can change throughout life as we learn, adapt and age. A balance between structural brain plasticity and homeostasis characterizes the healthy brain, and the breakdown of this balance accompanies brain tumors, psychiatric disorders, and neurodegenerative diseases. However, the link between circuit modifications, brain function, and behavior remains unclear. Importantly, the underlying molecular mechanisms are starting to be uncovered. The fruit-fly Drosophila is a very powerful model organism to discover molecular mechanisms and test them in vivo. There is abundant evidence that the Drosophila brain is plastic, and here we travel from the pioneering discoveries to recent findings and progress on molecular mechanisms. We pause on the recent discovery that, in the Drosophila central nervous system, Toll receptors—which bind neurotrophin ligands—regulate structural plasticity during development and in the adult brain. Through their topographic distribution across distinct brain modules and their ability to switch between alternative signaling outcomes, Tolls can enable the brain to translate experience into structural change. Intriguing similarities between Toll and mammalian Toll-like receptor function could reveal a further involvement in structural plasticity, degeneration, and disease in the human brain.
Collapse
Affiliation(s)
- Guiyi Li
- Plasticity and Regeneration Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Alicia Hidalgo
- Plasticity and Regeneration Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
10
|
Losada-Pérez M, García-Guillén N, Casas-Tintó S. A novel injury paradigm in the central nervous system of adult Drosophila: molecular, cellular and functional aspects. Dis Model Mech 2021; 14:268374. [PMID: 34061177 PMCID: PMC8214735 DOI: 10.1242/dmm.044669] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 03/24/2021] [Indexed: 11/24/2022] Open
Abstract
The mammalian central nervous system (CNS) exhibits limited regenerative capacity and the mechanisms that mediate its regeneration are not fully understood. Here, we present a novel experimental design to damage the CNS by using a contusion injury paradigm. The design of this protocol allows the study of long-term and short-term cellular responses, including those of the CNS and the immune system, and of any implications regarding functional recovery. We demonstrate for the first time that adult Drosophilamelanogaster glial cells undergo spontaneous functional recovery following crush injury. This crush injury leads to an intermediate level of functional recovery after damage, which is ideal to screen for genes that facilitate or prevent the regeneration process. Here, we validate this model and analyse the immune responses of glial cells as a central regulator of functional regeneration. Additionally, we demonstrate that glial cells and macrophages contribute to functional regeneration through mechanisms involving the Jun N-terminal kinase (JNK) pathway and the Drosophila protein Draper (Drpr), characteristic of other neural injury paradigms. We show that macrophages are recruited to the injury site and are required for functional recovery. Further, we show that the proteins Grindelwald and Drpr in Drosophila glial cells mediate activation of JNK, and that expression of drpr is dependent on JNK activation. Finally, we link neuron-glial communication and the requirement of neuronal vesicular transport to regulation of the JNK pathway and functional recovery. This article has an associated First Person interview with the first author of the paper. Summary: Central nervous system crush injury paradigm in adult Drosophilamelanogaster is a suitable model to study the cellular events, and genetic pathways behind injury responses and functional regeneration. We describe the immune responses of glial cells, neurons and macrophages following injury, and the functional relevance of each response.
Collapse
Affiliation(s)
- María Losada-Pérez
- Instituto Cajal-CSIC, Department of Molecular, Cellular and Developmental Neurobiology, 28002 Madrid, Spain
| | - Nuria García-Guillén
- Instituto Cajal-CSIC, Department of Molecular, Cellular and Developmental Neurobiology, 28002 Madrid, Spain
| | - Sergio Casas-Tintó
- Instituto Cajal-CSIC, Department of Molecular, Cellular and Developmental Neurobiology, 28002 Madrid, Spain
| |
Collapse
|
11
|
Mangold CA, Hughes DP. Insect Behavioral Change and the Potential Contributions of Neuroinflammation-A Call for Future Research. Genes (Basel) 2021; 12:465. [PMID: 33805190 PMCID: PMC8064348 DOI: 10.3390/genes12040465] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/21/2022] Open
Abstract
Many organisms are able to elicit behavioral change in other organisms. Examples include different microbes (e.g., viruses and fungi), parasites (e.g., hairworms and trematodes), and parasitoid wasps. In most cases, the mechanisms underlying host behavioral change remain relatively unclear. There is a growing body of literature linking alterations in immune signaling with neuron health, communication, and function; however, there is a paucity of data detailing the effects of altered neuroimmune signaling on insect neuron function and how glial cells may contribute toward neuron dysregulation. It is important to consider the potential impacts of altered neuroimmune communication on host behavior and reflect on its potential role as an important tool in the "neuro-engineer" toolkit. In this review, we examine what is known about the relationships between the insect immune and nervous systems. We highlight organisms that are able to influence insect behavior and discuss possible mechanisms of behavioral manipulation, including potentially dysregulated neuroimmune communication. We close by identifying opportunities for integrating research in insect innate immunity, glial cell physiology, and neurobiology in the investigation of behavioral manipulation.
Collapse
Affiliation(s)
- Colleen A. Mangold
- Department of Entomology, College of Agricultural Sciences, Pennsylvania State University, University Park, State College, PA 16802, USA;
- Center for Infectious Disease Dynamics, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, State College, PA 16802, USA
| | - David P. Hughes
- Department of Entomology, College of Agricultural Sciences, Pennsylvania State University, University Park, State College, PA 16802, USA;
- Center for Infectious Disease Dynamics, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, State College, PA 16802, USA
- Department of Biology, Eberly College of Science, Pennsylvania State University, University Park, State College, PA 16802, USA
| |
Collapse
|
12
|
Fernández-Hernández I, Marsh EB, Bonaguidi MA. Mechanosensory neuron regeneration in adult Drosophila. Development 2021; 148:dev.187534. [PMID: 33597190 DOI: 10.1242/dev.187534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
Auditory and vestibular mechanosensory hair cells do not regenerate following injury or aging in the adult mammalian inner ear, inducing irreversible hearing loss and balance disorders for millions of people. Research on model systems showing replacement of mechanosensory cells can provide mechanistic insights into developing new regenerative therapies. Here, we developed lineage tracing systems to reveal the generation of mechanosensory neurons in the Johnston's organ (JO) of intact adult Drosophila, which are the functional counterparts to hair cells in vertebrates. New JO neurons develop cilia and target central brain circuitry. Unexpectedly, mitotic recombination clones point to JO neuron self-replication as a likely source of neuronal plasticity. This mechanism is further enhanced upon treatment with experimental and ototoxic compounds. Our findings introduce a new platform to expedite research on mechanisms and compounds mediating mechanosensory cell regeneration, with nascent implications for hearing and balance restoration.
Collapse
Affiliation(s)
- Ismael Fernández-Hernández
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Evan B Marsh
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Michael A Bonaguidi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA .,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Gerontology, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033 USA
| |
Collapse
|
13
|
Kessissoglou IA, Langui D, Hasan A, Maral M, Dutta SB, Hiesinger PR, Hassan BA. The Drosophila amyloid precursor protein homologue mediates neuronal survival and neuroglial interactions. PLoS Biol 2020; 18:e3000703. [PMID: 33290404 PMCID: PMC7723294 DOI: 10.1371/journal.pbio.3000703] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022] Open
Abstract
The amyloid precursor protein (APP) is a structurally and functionally conserved transmembrane protein whose physiological role in adult brain function and health is still unclear. Because mutations in APP cause familial Alzheimer's disease (fAD), most research focuses on this aspect of APP biology. We investigated the physiological function of APP in the adult brain using the fruit fly Drosophila melanogaster, which harbors a single APP homologue called APP Like (APPL). Previous studies have provided evidence for the implication of APPL in neuronal wiring and axonal growth through the Wnt signaling pathway during development. However, like APP, APPL continues to be expressed in all neurons of the adult brain where its functions and their molecular and cellular underpinnings are unknown. We report that APPL loss of function (LOF) results in the dysregulation of endolysosomal function in neurons, with a notable enlargement of early endosomal compartments followed by neuronal cell death and the accumulation of dead neurons in the brain during a critical period at a young age. These defects can be rescued by reduction in the levels of the early endosomal regulator Rab5, indicating a causal role of endosomal function for cell death. Finally, we show that the secreted extracellular domain of APPL interacts with glia and regulates the size of their endosomes, the expression of the Draper engulfment receptor, and the clearance of neuronal debris in an axotomy model. We propose that APP proteins represent a novel family of neuroglial signaling factors required for adult brain homeostasis.
Collapse
Affiliation(s)
- Irini A. Kessissoglou
- Paris Brain Institute, Hôpital Pitié-Salpêtrière, Inserm U 1127, CNRS UMR, Sorbonne Université, Paris, France
| | - Dominique Langui
- Paris Brain Institute, Hôpital Pitié-Salpêtrière, Inserm U 1127, CNRS UMR, Sorbonne Université, Paris, France
| | - Amr Hasan
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Maral Maral
- Paris Brain Institute, Hôpital Pitié-Salpêtrière, Inserm U 1127, CNRS UMR, Sorbonne Université, Paris, France
| | - Suchetana B. Dutta
- Paris Brain Institute, Hôpital Pitié-Salpêtrière, Inserm U 1127, CNRS UMR, Sorbonne Université, Paris, France
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Peter Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Bassem A. Hassan
- Paris Brain Institute, Hôpital Pitié-Salpêtrière, Inserm U 1127, CNRS UMR, Sorbonne Université, Paris, France
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
14
|
Khor S, Cai D. Control of lifespan and survival by Drosophila NF-κB signaling through neuroendocrine cells and neuroblasts. Aging (Albany NY) 2020; 12:24604-24622. [PMID: 33232282 PMCID: PMC7803524 DOI: 10.18632/aging.104196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022]
Abstract
We report a comparative analysis of the effects of immune activation in the fly nervous system using genetic activation models to target Drosophila NF-κB within Toll versus Imd pathways. Genetic gain-of-function models for either pathway pan-neuronally as well as in discrete subsets of neural cells including neuroendocrine insulin-producing cells (IPCs) or neuroblasts reduce fly lifespan, however, these phenotypes in IPCs and neuroblasts are stronger with Toll activation than Imd activation. Of note, while aging is influenced more by Toll/NF-κB activation in IPCs during adulthood, neuroblasts influence aging more substantially during development. The study then focused on Toll/NF-κB inhibition, revealing that IPCs or neuroblasts are important for the effects of lifespan and healthspan extension but in a life stage-dependent manner while some of these effects display sexual dimorphism. Importantly, co-inhibition of Toll/NF-κB pathway in IPCs and neuroblasts increased fly lifespan greater than either cell population, suggesting that independent mechanisms might exist. Toll/NF-κB inhibition in IPCs was also sufficient to enhance survival under various fatal stresses, supporting the additional benefits to fly healthspan. In conclusion, IPCs and neuroblasts are important for Drosophila NF-κB for controlling lifespan.
Collapse
Affiliation(s)
- Sinan Khor
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
15
|
Li G, Hidalgo A. Adult Neurogenesis in the Drosophila Brain: The Evidence and the Void. Int J Mol Sci 2020; 21:ijms21186653. [PMID: 32932867 PMCID: PMC7554932 DOI: 10.3390/ijms21186653] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 01/05/2023] Open
Abstract
Establishing the existence and extent of neurogenesis in the adult brain throughout the animals including humans, would transform our understanding of how the brain works, and how to tackle brain damage and disease. Obtaining convincing, indisputable experimental evidence has generally been challenging. Here, we revise the state of this question in the fruit-fly Drosophila. The developmental neuroblasts that make the central nervous system and brain are eliminated, either through apoptosis or cell cycle exit, before the adult fly ecloses. Despite this, there is growing evidence that cell proliferation can take place in the adult brain. This occurs preferentially at, but not restricted to, a critical period. Adult proliferating cells can give rise to both glial cells and neurons. Neuronal activity, injury and genetic manipulation in the adult can increase the incidence of both gliogenesis and neurogenesis, and cell number. Most likely, adult glio- and neuro-genesis promote structural brain plasticity and homeostasis. However, a definitive visualisation of mitosis in the adult brain is still lacking, and the elusive adult progenitor cells are yet to be identified. Resolving these voids is important for the fundamental understanding of any brain. Given its powerful genetics, Drosophila can expedite discovery into mammalian adult neurogenesis in the healthy and diseased brain.
Collapse
|
16
|
Nandakumar S, Grushko O, Buttitta LA. Polyploidy in the adult Drosophila brain. eLife 2020; 9:e54385. [PMID: 32840209 PMCID: PMC7447450 DOI: 10.7554/elife.54385] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Long-lived cells such as terminally differentiated postmitotic neurons and glia must cope with the accumulation of damage over the course of an animal's lifespan. How long-lived cells deal with ageing-related damage is poorly understood. Here we show that polyploid cells accumulate in the adult fly brain and that polyploidy protects against DNA damage-induced cell death. Multiple types of neurons and glia that are diploid at eclosion, become polyploid in the adult Drosophila brain. The optic lobes exhibit the highest levels of polyploidy, associated with an elevated DNA damage response in this brain region. Inducing oxidative stress or exogenous DNA damage leads to an earlier onset of polyploidy, and polyploid cells in the adult brain are more resistant to DNA damage-induced cell death than diploid cells. Our results suggest polyploidy may serve a protective role for neurons and glia in adult Drosophila melanogaster brains.
Collapse
Affiliation(s)
- Shyama Nandakumar
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Olga Grushko
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| | - Laura A Buttitta
- Molecular, Cellular, and Developmental Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
17
|
Sheng L, Shields EJ, Gospocic J, Glastad KM, Ratchasanmuang P, Berger SL, Raj A, Little S, Bonasio R. Social reprogramming in ants induces longevity-associated glia remodeling. SCIENCE ADVANCES 2020; 6:eaba9869. [PMID: 32875108 PMCID: PMC7438095 DOI: 10.1126/sciadv.aba9869] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/09/2020] [Indexed: 05/16/2023]
Abstract
In social insects, workers and queens arise from the same genome but display profound differences in behavior and longevity. In Harpegnathos saltator ants, adult workers can transition to a queen-like state called gamergate, which results in reprogramming of social behavior and life-span extension. Using single-cell RNA sequencing, we compared the distribution of neuronal and glial populations before and after the social transition. We found that the conversion of workers into gamergates resulted in the expansion of neuroprotective ensheathing glia. Brain injury assays revealed that activation of the damage response gene Mmp1 was weaker in old workers, where the relative frequency of ensheathing glia also declined. On the other hand, long-lived gamergates retained a larger fraction of ensheathing glia and the ability to mount a strong Mmp1 response to brain injury into old age. We also observed molecular and cellular changes suggestive of age-associated decline in ensheathing glia in Drosophila.
Collapse
Affiliation(s)
- Lihong Sheng
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emily J. Shields
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Janko Gospocic
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karl M. Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Puttachai Ratchasanmuang
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shelley L. Berger
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arjun Raj
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Shawn Little
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Roberto Bonasio
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Upregulated TNF/Eiger signaling mediates stem cell recovery and tissue homeostasis during nutrient resupply in Drosophila testis. Sci Rep 2020; 10:11674. [PMID: 32669615 PMCID: PMC7363678 DOI: 10.1038/s41598-020-68313-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Stem cell activity and cell differentiation is robustly influenced by the nutrient availability in the gonads. The signal that connects nutrient availability to gonadal stem cell activity remains largely unknown. In this study, we show that tumor necrosis factor Eiger (Egr) is upregulated in testicular smooth muscles as a response to prolonged protein starvation in Drosophila testis. While Egr is not essential for starvation-induced changes in germline and somatic stem cell numbers, Egr and its receptor Grindelwald influence the recovery dynamics of somatic cyst stem cells (CySCs) upon protein refeeding. Moreover, Egr is also involved in the refeeding-induced, ectopic expression of the CySC self-renewal protein and the accumulation of early germ cells. Egr primarily acts through the Jun N-terminal kinase (JNK) signaling in Drosophila. We show that inhibition of JNK signaling in cyst cells suppresses the refeeding-induced abnormality in both somatic and germ cells. In conclusion, our study reveals both beneficial and detrimental effects of Egr upregulation in the recovery of stem cells and spermatogenesis from prolonged protein starvation.
Collapse
|
19
|
Kato K, Orihara-Ono M, Awasaki T. Multiple lineages enable robust development of the neuropil-glia architecture in adult Drosophila. Development 2020; 147:dev184085. [PMID: 32051172 DOI: 10.1242/dev.184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Neural remodeling is essential for the development of a functional nervous system and has been extensively studied in the metamorphosis of Drosophila Despite the crucial roles of glial cells in brain functions, including learning and behavior, little is known of how adult glial cells develop in the context of neural remodeling. Here, we show that the architecture of neuropil-glia in the adult Drosophila brain, which is composed of astrocyte-like glia (ALG) and ensheathing glia (EG), robustly develops from two different populations in the larva: the larval EG and glial cell missing-positive (gcm+ ) cells. Whereas gcm+ cells proliferate and generate adult ALG and EG, larval EG dedifferentiate, proliferate and redifferentiate into the same glial subtypes. Each glial lineage occupies a certain brain area complementary to the other, and together they form the adult neuropil-glia architecture. Both lineages require the FGF receptor Heartless to proliferate, and the homeoprotein Prospero to differentiate into ALG. Lineage-specific inhibition of gliogenesis revealed that each lineage compensates for deficiency in the proliferation of the other. Together, the lineages ensure the robust development of adult neuropil-glia, thereby ensuring a functional brain.
Collapse
Affiliation(s)
- Kentaro Kato
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Minako Orihara-Ono
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Takeshi Awasaki
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| |
Collapse
|
20
|
Li G, Forero MG, Wentzell JS, Durmus I, Wolf R, Anthoney NC, Parker M, Jiang R, Hasenauer J, Strausfeld NJ, Heisenberg M, Hidalgo A. A Toll-receptor map underlies structural brain plasticity. eLife 2020; 9:52743. [PMID: 32066523 PMCID: PMC7077983 DOI: 10.7554/elife.52743] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/12/2020] [Indexed: 12/28/2022] Open
Abstract
Experience alters brain structure, but the underlying mechanism remained unknown. Structural plasticity reveals that brain function is encoded in generative changes to cells that compete with destructive processes driving neurodegeneration. At an adult critical period, experience increases fiber number and brain size in Drosophila. Here, we asked if Toll receptors are involved. Tolls demarcate a map of brain anatomical domains. Focusing on Toll-2, loss of function caused apoptosis, neurite atrophy and impaired behaviour. Toll-2 gain of function and neuronal activity at the critical period increased cell number. Toll-2 induced cycling of adult progenitor cells via a novel pathway, that antagonized MyD88-dependent quiescence, and engaged Weckle and Yorkie downstream. Constant knock-down of multiple Tolls synergistically reduced brain size. Conditional over-expression of Toll-2 and wek at the adult critical period increased brain size. Through their topographic distribution, Toll receptors regulate neuronal number and brain size, modulating structural plasticity in the adult brain. Everything that you experience leaves its mark on your brain. When you learn something new, the neurons involved in the learning episode grow new projections and form new connections. Your brain may even produce new neurons. Physical exercise can induce similar changes, as can taking antidepressants. By contrast, stress, depression, ageing and disease can have the opposite effect, triggering neurons to break down and even die. The ability of the brain to change in response to experience is known as structural plasticity, and it is in a tug-of-war with processes that drive neurodegeneration. Structural plasticity occurs in other species too: for example, it was described in the fruit fly more than a quarter of a century ago. Yet, the molecular mechanisms underlying structural plasticity remain unclear. Li et al. now show that, in fruit flies, this plasticity involves Toll receptors, a family of proteins present in the brain but best known for their role in the immune system. Fruit flies have nine different Toll receptors, the most abundant being Toll-2. When activated, these proteins can trigger a series of molecular events in a cell. Li et al. show that increasing the amount of Toll-2 in the fly brain makes the brain produce new neurons. Activating neurons in a brain region has the same effect, and this increase in neuron number also depends on Toll-2. By contrast, reducing the amount of Toll-2 causes neurons to lose their projections and connections, and to die, and impairs fly behaviour. Li et al. also show that each Toll receptor has a unique distribution across the fly brain. Different types of experiences activate different brain regions, and therefore different Toll receptors. These go on to trigger a common molecular cascade, but they modulate it such as to result in distinct outcomes. By working together in different combinations, Toll receptors can promote either the death or survival of neurons, and they can also drive specific brain cells to remain dormant or to produce new neurons. By revealing how experience changes the brain, Li et al. provide clues to the way neurons work and form; these findings may also help to find new treatments for disorders that change brain structure, such as certain psychiatric conditions. Toll-like receptors in humans could thus represent a promising new target for drug discovery.
Collapse
Affiliation(s)
- Guiyi Li
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom.,Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Manuel G Forero
- Facultad de Ingeniería, Universidad de Ibagué, Ibagué, Colombia
| | - Jill S Wentzell
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Ilgim Durmus
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Reinhard Wolf
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Niki C Anthoney
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Mieczyslaw Parker
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Ruiying Jiang
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Jacob Hasenauer
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Nicholas James Strausfeld
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany.,Neuroscience, University of Arizona College of Science, Tucson, United States
| | - Martin Heisenberg
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Alicia Hidalgo
- Neurodevelopment Lab, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
21
|
Vásquez-Procopio J, Osorio B, Cortés-Martínez L, Hernández-Hernández F, Medina-Contreras O, Ríos-Castro E, Comjean A, Li F, Hu Y, Mohr S, Perrimon N, Missirlis F. Intestinal response to dietary manganese depletion inDrosophila. Metallomics 2020; 12:218-240. [DOI: 10.1039/c9mt00218a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metabolic adaptations to manganese deficiency.
Collapse
|
22
|
van den Ameele J, Brand AH. Neural stem cell temporal patterning and brain tumour growth rely on oxidative phosphorylation. eLife 2019; 8:47887. [PMID: 31513013 PMCID: PMC6763261 DOI: 10.7554/elife.47887] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Translating advances in cancer research to clinical applications requires better insight into the metabolism of normal cells and tumour cells in vivo. Much effort has focused on understanding how glycolysis and oxidative phosphorylation (OxPhos) support proliferation, while their impact on other aspects of development and tumourigenesis remain largely unexplored. We found that inhibition of OxPhos in neural stem cells (NSCs) or tumours in the Drosophila brain not only decreases proliferation, but also affects many different aspects of stem cell behaviour. In NSCs, OxPhos dysfunction leads to a protracted G1/S-phase and results in delayed temporal patterning and reduced neuronal diversity. As a consequence, NSCs fail to undergo terminal differentiation, leading to prolonged neurogenesis into adulthood. Similarly, in brain tumours inhibition of OxPhos slows proliferation and prevents differentiation, resulting in reduced tumour heterogeneity. Thus, in vivo, highly proliferative stem cells and tumour cells require OxPhos for efficient growth and generation of diversity.
Collapse
Affiliation(s)
- Jelle van den Ameele
- The Gurdon Institute, Cambridge, United Kingdom.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andrea H Brand
- The Gurdon Institute, Cambridge, United Kingdom.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
23
|
Melcarne C, Lemaitre B, Kurant E. Phagocytosis in Drosophila: From molecules and cellular machinery to physiology. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 109:1-12. [PMID: 30953686 DOI: 10.1016/j.ibmb.2019.04.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/22/2019] [Accepted: 04/01/2019] [Indexed: 05/20/2023]
Abstract
Phagocytosis is an evolutionarily conserved mechanism that plays a key role in both host defence and tissue homeostasis in multicellular organisms. A range of surface receptors expressed on different cell types allow discriminating between self and non-self (or altered) material, thus enabling phagocytosis of pathogens and apoptotic cells. The phagocytosis process can be divided into four main steps: 1) binding of the phagocyte to the target particle, 2) particle internalization and phagosome formation, through remodelling of the plasma membrane, 3) phagosome maturation, and 4) particle destruction in the phagolysosome. In this review, we describe our present knowledge on phagocytosis in the fruit fly Drosophila melanogaster, assessing each of the key steps involved in engulfment of both apoptotic cells and bacteria. We also assess the physiological role of phagocytosis in host defence, development and tissue homeostasis.
Collapse
Affiliation(s)
- C Melcarne
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - B Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - E Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, 34988, Israel.
| |
Collapse
|
24
|
E93 Integrates Neuroblast Intrinsic State with Developmental Time to Terminate MB Neurogenesis via Autophagy. Curr Biol 2019; 29:750-762.e3. [PMID: 30773368 DOI: 10.1016/j.cub.2019.01.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/05/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Most neurogenesis occurs during development, driven by the cell divisions of neural stem cells (NSCs). We use Drosophila to understand how neurogenesis terminates once development is complete, a process critical for neural circuit formation. We identified E93, a steroid-hormone-induced transcription factor that downregulates phosphatidylinositol 3-kinase (PI3K) levels to activate autophagy for elimination of mushroom body (MB) neuroblasts. MB neuroblasts are a subset of Drosophila NSCs that generate neurons important for memory and learning. MB neurogenesis extends into adulthood when E93 is reduced and terminates prematurely when E93 is overexpressed. E93 is expressed in MB neuroblasts during later stages of pupal development only, which includes the time when MB neuroblasts normally terminate their divisions. Cell intrinsic Imp and Syp temporal factors regulate timing of E93 expression in MB neuroblasts, and extrinsic steroid hormone receptor (EcR) activation boosts E93 levels high for termination. Imp inhibits premature expression of E93 in a Syp-dependent manner, and Syp positively regulates E93 to promote neurogenesis termination. Imp and Syp together with E93 form a temporal cassette, which consequently links early developmental neurogenesis with termination. Altogether, E93 functions as a late-acting temporal factor integrating extrinsic hormonal cues linked to developmental timing with neuroblast intrinsic temporal cues to precisely time neurogenesis ending during development.
Collapse
|
25
|
Losada-Perez M. Glia: from 'just glue' to essential players in complex nervous systems: a comparative view from flies to mammals. J Neurogenet 2018; 32:78-91. [PMID: 29718753 DOI: 10.1080/01677063.2018.1464568] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the last years, glial cells have emerged as central players in the development and function of complex nervous systems. Therefore, the concept of glial cells has evolved from simple supporting cells to essential actors. The molecular mechanisms that govern glial functions are evolutionarily conserved from Drosophila to mammals, highlighting genetic similarities between these groups, as well as the great potential of Drosophila research for the understanding of human CNS. These similarities would imply a common phylogenetic origin of glia, even though there is a controversy at this point. This review addresses the existing literature on the evolutionary origin of glia and discusses whether or not insect and mammalian glia are homologous or analogous. Besides, this manuscript summarizes the main glial functions in the CNS and underscores the evolutionarily conserved molecular mechanisms between Drosophila and mammals. Finally, I also consider the current nomenclature and classification of glial cells to highlight the need for a consensus agreement and I propose an alternative nomenclature based on function that unifies Drosophila and mammalian glial types.
Collapse
|
26
|
When dying is not the end: Apoptotic caspases as drivers of proliferation. Semin Cell Dev Biol 2017; 82:86-95. [PMID: 29199139 DOI: 10.1016/j.semcdb.2017.11.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022]
Abstract
Caspases are well known for their role as executioners of apoptosis. However, recent studies have revealed that these lethal enzymes also have important mitogenic functions. Caspases can promote proliferation through autonomous regulation of the cell cycle, as well as by induction of secreted signals, which have a profound impact in neighboring tissues. Here, I review the proliferative role of caspases during development and homeostasis, in addition to their key regenerative function during tissue repair upon injury. Furthermore, the emerging properties of apoptotic caspases as drivers of carcinogenesis are discussed, as well as their involvement in other diseases. Finally, I examine further effects of caspases regulating death and survival in a non-autonomous manner.
Collapse
|
27
|
Go and stop signals for glial regeneration. Curr Opin Neurobiol 2017; 47:182-187. [PMID: 29126016 PMCID: PMC6419527 DOI: 10.1016/j.conb.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/16/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
The regenerative response of ensheating glia to central nervous system (CNS) injury involves proliferation and differentiation, axonal re-enwrapment and some recovery of behaviour. Understanding this limited response could enable the enhancement of it. In Drosophila, the glial progenitor state is maintained by Notch, an activator of cell division and Prospero (Pros), a repressor. Injury provokes the activation of NFκB and up-regulation of Kon-tiki (Kon), driving cell proliferation. Homeostatic switch-off comes about as two negative feedback loops involving Pros terminate the response. Importantly, the functions of the kon and pros homologues NG2 and prox1, respectively, are conserved in mammalian NG2 glia. Controlling these genes is key for therapeutic manipulation of progenitors and stem cells to promote regeneration of the damaged CNS.
Collapse
|
28
|
Kato K, Losada-Perez M, Hidalgo A. Gene network underlying the glial regenerative response to central nervous system injury. Dev Dyn 2017; 247:85-93. [DOI: 10.1002/dvdy.24565] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Kentaro Kato
- School of Medicine; Kyorin University; Tokyo Japan
| | | | - Alicia Hidalgo
- School of Biosciences; University of Birmingham; United Kingdom
| |
Collapse
|
29
|
Losick VP. Wound-Induced Polyploidy Is Required for Tissue Repair. Adv Wound Care (New Rochelle) 2016; 5:271-278. [PMID: 27274437 DOI: 10.1089/wound.2014.0545] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Significance: All organs suffer wounds to some extent during an animal's lifetime and to compensate for cell loss, tissues often rely on cell division. However, many organs are made up of differentiated cells with only a limited capacity to divide. It is not well understood how cells are replaced in the absence of cell division. Recent Advances: Recent studies in the model organism Drosophila melanogaster have proven that wound-induced polyploidy (WIP) is an essential mechanism to replace tissue mass and restore tissue integrity in the absence of cell division. In this repair mechanism, preexisting differentiated cells increase their DNA content and cell size by becoming polyploid. Critical Issues: Cells within mammalian organs such as the liver, heart, and cornea have also been observed to increase their DNA ploidy in response to injury, suggesting that WIP may be an evolutionarily conserved mechanism to compensate for cell loss. Future Directions: The Hippo signal transduction pathway is required for differentiated cells to initiate WIP in Drosophila. Continued studies in Drosophila will help to identify other signaling pathways required for WIP as well as the conserved mechanisms that polyploid cells may play during wound repair in all organisms.
Collapse
Affiliation(s)
- Vicki P. Losick
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| |
Collapse
|
30
|
Melzer J, Broemer M. Nerve-racking - apoptotic and non-apoptotic roles of caspases in the nervous system of Drosophila. Eur J Neurosci 2016; 44:1683-90. [PMID: 26900934 DOI: 10.1111/ejn.13213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 02/02/2016] [Accepted: 02/15/2016] [Indexed: 12/28/2022]
Abstract
Studies using Drosophila as a model system have contributed enormously to our knowledge of caspase function and regulation. Caspases are best known as central executioners of apoptosis but also control essential physiological processes in a non-apoptotic manner. The Drosophila genome codes for seven caspases and in this review we provide an overview of current knowledge about caspase function in the nervous system. Caspases regulate neuronal death at all developmental stages and in various neuronal populations. In contrast, non-apoptotic roles are less well understood. The development of new genetically encoded sensors for caspase activity provides unprecedented opportunities to study caspase function in the nervous system in more detail. In light of these new tools we discuss the potential of Drosophila as a model to discover new apoptotic and non-apoptotic neuronal roles of caspases.
Collapse
Affiliation(s)
- Juliane Melzer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Meike Broemer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
31
|
Pérez-Garijo A, Steller H. Spreading the word: non-autonomous effects of apoptosis during development, regeneration and disease. Development 2016; 142:3253-62. [PMID: 26443630 DOI: 10.1242/dev.127878] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apoptosis, in contrast to other forms of cell death such as necrosis, was originally regarded as a 'silent' mechanism of cell elimination designed to degrade the contents of doomed cells. However, during the past decade it has become clear that apoptotic cells can produce diverse signals that have a profound impact on neighboring cells and tissues. For example, apoptotic cells can release factors that influence the proliferation and survival of adjacent tissues. Apoptosis can also affect tissue movement and morphogenesis by modifying tissue tension in surrounding cells. As we review here, these findings reveal unexpected roles for apoptosis in tissue remodeling during development, as well as in regeneration and cancer.
Collapse
Affiliation(s)
- Ainhoa Pérez-Garijo
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
32
|
Panov AA. Prolonged persistence of neuroblast clusters in the optic lobes of the ground beetles Carabus nemoralis Müll. and C. hortensis L. (Coleoptera: Carabidae). BIOL BULL+ 2016. [DOI: 10.1134/s106235901601012x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Yasugi T, Nishimura T. Temporal regulation of the generation of neuronal diversity in Drosophila. Dev Growth Differ 2015; 58:73-87. [PMID: 26690868 DOI: 10.1111/dgd.12245] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/10/2015] [Accepted: 10/10/2015] [Indexed: 12/18/2022]
Abstract
For the construction of complex neural networks, the generation of neurons and glia must be tightly regulated both spatially and temporally. One of the major issues in neural development is the generation of a large variety of neurons and glia over time from a relatively small number of neural stem cells. In Drosophila, neural stem cells, called neuroblasts (NBs), have been used as a useful model system to uncover the molecular and cellular machinery involved in the establishment of neural diversity. NBs divide asymmetrically and produce another self-renewing progenitor cell and a differentiating cell. NBs are subdivided into several types based on their location in the central nervous system. Each type of NB has specific features related to the timing of cell generation, cell cycle progression, temporal patterning for neuronal specification, and termination mechanism. In this review, we focus on the molecular mechanisms that regulate the proliferation of NBs and generate a large variety of neuronal and glia subtypes during development.
Collapse
Affiliation(s)
- Tetsuo Yasugi
- Laboratory for Growth Control Signaling, RIKEN Center for Developmental Biology (CDB), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takashi Nishimura
- Laboratory for Growth Control Signaling, RIKEN Center for Developmental Biology (CDB), 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
34
|
Dheilly NM, Maure F, Ravallec M, Galinier R, Doyon J, Duval D, Leger L, Volkoff AN, Missé D, Nidelet S, Demolombe V, Brodeur J, Gourbal B, Thomas F, Mitta G. Who is the puppet master? Replication of a parasitic wasp-associated virus correlates with host behaviour manipulation. Proc Biol Sci 2015; 282:20142773. [PMID: 25673681 DOI: 10.1098/rspb.2014.2773] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Many parasites modify their host behaviour to improve their own transmission and survival, but the proximate mechanisms remain poorly understood. An original model consists of the parasitoid Dinocampus coccinellae and its coccinellid host, Coleomegilla maculata; during the behaviour manipulation, the parasitoid is not in contact with its host anymore. We report herein the discovery and characterization of a new RNA virus of the parasitoid (D. coccinellae paralysis virus, DcPV). Using a combination of RT-qPCR and transmission electron microscopy, we demonstrate that DcPV is stored in the oviduct of parasitoid females, replicates in parasitoid larvae and is transmitted to the host during larval development. Next, DcPV replication in the host's nervous tissue induces a severe neuropathy and antiviral immune response that correlate with the paralytic symptoms characterizing the behaviour manipulation. Remarkably, virus clearance correlates with recovery of normal coccinellid behaviour. These results provide evidence that changes in ladybeetle behaviour most likely result from DcPV replication in the cerebral ganglia rather than by manipulation by the parasitoid. This offers stimulating prospects for research on parasitic manipulation by suggesting for the first time that behaviour manipulation could be symbiont-mediated.
Collapse
Affiliation(s)
- Nolwenn M Dheilly
- UMR 5244, Ecologie et Evolution des Interactions (2EI), CNRS, Université de Perpignan, Perpignan 66860, France MIVEGEC (UMR CNRS/IRD/UM1/UM2 5290), 911 Avenue Agropolis, BP 64501, Montpellier Cedex 5 34394, France
| | - Fanny Maure
- MIVEGEC (UMR CNRS/IRD/UM1/UM2 5290), 911 Avenue Agropolis, BP 64501, Montpellier Cedex 5 34394, France Département de Sciences Biologiques, Institut de Recherche en Biologie Végétale, Université de Montréal, 4101 rue Sherbrooke est, Montréal, Québec, Canada H1X 2B2
| | - Marc Ravallec
- INRA (UMR 1333), 'Insect-Microorganisms Diversity, Genomes and Interactions', Université de Montpellier 2, Place Eugène Bataillon, CC101, Montpellier Cedex 34095, France
| | - Richard Galinier
- UMR 5244, Ecologie et Evolution des Interactions (2EI), CNRS, Université de Perpignan, Perpignan 66860, France
| | - Josée Doyon
- Département de Sciences Biologiques, Institut de Recherche en Biologie Végétale, Université de Montréal, 4101 rue Sherbrooke est, Montréal, Québec, Canada H1X 2B2
| | - David Duval
- UMR 5244, Ecologie et Evolution des Interactions (2EI), CNRS, Université de Perpignan, Perpignan 66860, France
| | - Lucas Leger
- MIVEGEC (UMR CNRS/IRD/UM1/UM2 5290), 911 Avenue Agropolis, BP 64501, Montpellier Cedex 5 34394, France
| | - Anne-Nathalie Volkoff
- INRA (UMR 1333), 'Insect-Microorganisms Diversity, Genomes and Interactions', Université de Montpellier 2, Place Eugène Bataillon, CC101, Montpellier Cedex 34095, France
| | - Dorothée Missé
- MIVEGEC (UMR CNRS/IRD/UM1/UM2 5290), 911 Avenue Agropolis, BP 64501, Montpellier Cedex 5 34394, France
| | - Sabine Nidelet
- Montpellier Genomics and Bioinformatics Facility, MGX-Montpellier GenomiX, Montpellier 34396, France
| | - Vincent Demolombe
- Montpellier Genomics and Bioinformatics Facility, MGX-Montpellier GenomiX, Montpellier 34396, France
| | - Jacques Brodeur
- Département de Sciences Biologiques, Institut de Recherche en Biologie Végétale, Université de Montréal, 4101 rue Sherbrooke est, Montréal, Québec, Canada H1X 2B2
| | - Benjamin Gourbal
- UMR 5244, Ecologie et Evolution des Interactions (2EI), CNRS, Université de Perpignan, Perpignan 66860, France
| | - Frédéric Thomas
- MIVEGEC (UMR CNRS/IRD/UM1/UM2 5290), 911 Avenue Agropolis, BP 64501, Montpellier Cedex 5 34394, France
| | - Guillaume Mitta
- UMR 5244, Ecologie et Evolution des Interactions (2EI), CNRS, Université de Perpignan, Perpignan 66860, France
| |
Collapse
|
35
|
|
36
|
Fernández-Hernández I, Rhiner C. New neurons for injured brains? The emergence of new genetic model organisms to study brain regeneration. Neurosci Biobehav Rev 2015; 56:62-72. [PMID: 26118647 DOI: 10.1016/j.neubiorev.2015.06.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 11/15/2022]
Abstract
Neuronal circuits in the adult brain have long been viewed as static and stable. However, research in the past 20 years has shown that specialized regions of the adult brain, which harbor adult neural stem cells, continue to produce new neurons in a wide range of species. Brain plasticity is also observed after injury. Depending on the extent and permissive environment of neurogenic regions, different organisms show great variability in their capacity to replace lost neurons by endogenous neurogenesis. In Zebrafish and Drosophila, the formation of new neurons from progenitor cells in the adult brain was only discovered recently. Here, we compare properties of adult neural stem cells, their niches and regenerative responses from mammals to flies. Current models of brain injury have revealed that specific injury-induced genetic programs and comparison of neuronal fitness are implicated in brain repair. We highlight the potential of these recently implemented models of brain regeneration to identify novel regulators of stem cell activation and regenerative neurogenesis.
Collapse
Affiliation(s)
| | - Christa Rhiner
- Institute of Cell Biology, IZB, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|
37
|
Shklover J, Levy-Adam F, Kurant E. The role of Drosophila TNF Eiger in developmental and damage-induced neuronal apoptosis. FEBS Lett 2015; 589:871-9. [PMID: 25754009 DOI: 10.1016/j.febslet.2015.02.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/17/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022]
Abstract
Eiger, the sole Drosophila TNF-alpha homolog, causes ectopic apoptosis through JNK pathway activation. Yet, its role in developmental apoptosis remains unclear. eiger mutant flies are viable and fertile but display compromised elimination of oncogenic cells and extracellular bacteria. Here we show that Eiger, specifically expressed in embryonic neurons and glia, is not involved in developmental neuronal apoptosis or in apoptotic cell clearance. Instead, we provide evidence that Eiger is required for damage-induced apoptosis in the embryonic CNS through regulation of the pro-apoptotic gene hid independently of the JNK pathway. Our study thus reveals a new requirement for Eiger in eliminating damaged cells during development.
Collapse
Affiliation(s)
- Jeny Shklover
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Flonia Levy-Adam
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Estee Kurant
- Department of Genetics and Developmental Biology, The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
38
|
Glial enriched gene expression profiling identifies novel factors regulating the proliferation of specific glial subtypes in the Drosophila brain. Gene Expr Patterns 2014; 16:61-8. [PMID: 25217886 PMCID: PMC4222725 DOI: 10.1016/j.gep.2014.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 01/13/2023]
Abstract
Global gene expression analysis identifies glial specific transcriptomes. Different glial subtypes have distinct but overlapping transcriptomes. foxO and tramtrack69 are novel regulators of glial subtype specific proliferation.
Glial cells constitute a large proportion of the central nervous system (CNS) and are critical for the correct development and function of the adult CNS. Recent studies have shown that specific subtypes of glia are generated through the proliferation of differentiated glial cells in both the developing invertebrate and vertebrate nervous systems. However, the factors that regulate glial proliferation in specific glial subtypes are poorly understood. To address this we have performed global gene expression analysis of Drosophila post-embryonic CNS tissue enriched in glial cells, through glial specific overexpression of either the FGF or insulin receptor. Analysis of the differentially regulated genes in these tissues shows that the expression of known glial genes is significantly increased in both cases. Conversely, the expression of neuronal genes is significantly decreased. FGF and insulin signalling drive the expression of overlapping sets of genes in glial cells that then activate proliferation. We then used these data to identify novel transcription factors that are expressed in glia in the brain. We show that two of the transcription factors identified in the glial enriched gene expression profiles, foxO and tramtrack69, have novel roles in regulating the proliferation of cortex and perineurial glia. These studies provide new insight into the genes and molecular pathways that regulate the proliferation of specific glial subtypes in the Drosophila post-embryonic brain.
Collapse
|
39
|
Igaki T, Miura M. The Drosophila TNF ortholog Eiger: emerging physiological roles and evolution of the TNF system. Semin Immunol 2014; 26:267-74. [PMID: 24981286 DOI: 10.1016/j.smim.2014.05.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 05/22/2014] [Indexed: 12/19/2022]
Abstract
The TNF and TNFR superfamilies of proteins are conserved throughout evolution. The first invertebrate orthologs of TNF and TNFR, Eiger and Wengen, were identified in Drosophila, which enabled us to take advantage of its powerful genetics. Indeed, genetic studies on Eiger in the last decade have discovered their signaling mechanisms through activation of the JNK pathway and unveiled the role of Eiger-JNK signaling in a variety of cellular and tissue processes such as cell death, cell proliferation, tissue growth regulation, host defense, pain sensitization, and canalization. In this review, we will describe the in vivo signaling of Eiger and its physiological roles in fly development and homeostasis, and will discuss the evolution of the TNF/TNFR systems.
Collapse
Affiliation(s)
- Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoecho, Sakyo-ku, Kyoto 606-8501, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Basic Research Program, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0075, Japan.
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; Core Research for Evolutional Science and Technology (CREST), Basic Research Program, Japan Science and Technology Agency, Chiyoda-ku, Tokyo 102-0075, Japan.
| |
Collapse
|
40
|
Chihara T, Kitabayashi A, Morimoto M, Takeuchi KI, Masuyama K, Tonoki A, Davis RL, Wang JW, Miura M. Caspase inhibition in select olfactory neurons restores innate attraction behavior in aged Drosophila. PLoS Genet 2014; 10:e1004437. [PMID: 24967585 PMCID: PMC4072539 DOI: 10.1371/journal.pgen.1004437] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 04/27/2014] [Indexed: 01/31/2023] Open
Abstract
Sensory and cognitive performance decline with age. Neural dysfunction caused by nerve death in senile dementia and neurodegenerative disease has been intensively studied; however, functional changes in neural circuits during the normal aging process are not well understood. Caspases are key regulators of cell death, a hallmark of age-related neurodegeneration. Using a genetic probe for caspase-3-like activity (DEVDase activity), we have mapped age-dependent neuronal changes in the adult brain throughout the lifespan of Drosophila. Spatio-temporally restricted caspase activation was observed in the antennal lobe and ellipsoid body, brain structures required for olfaction and visual place memory, respectively. We also found that caspase was activated in an age-dependent manner in specific subsets of Drosophila olfactory receptor neurons (ORNs), Or42b and Or92a neurons. These neurons are essential for mediating innate attraction to food-related odors. Furthermore, age-induced impairments of neural transmission and attraction behavior could be reversed by specific inhibition of caspase in these ORNs, indicating that caspase activation in Or42b and Or92a neurons is responsible for altering animal behavior during normal aging.
Collapse
Affiliation(s)
- Takahiro Chihara
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency (JST), Gobancho, Chiyoda-ku, Tokyo, Japan
- CREST, Japan Science and Technology Agency (JST), Gobancho, Chiyoda-ku, Tokyo, Japan
- * E-mail: (TC); (MMi)
| | - Aki Kitabayashi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Michie Morimoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Ken-ichi Takeuchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kaoru Masuyama
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Ayako Tonoki
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, Florida, United States of America
- Department of Biochemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana, Chuo-ku, Chiba, Japan
| | - Ronald L. Davis
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, Florida, United States of America
| | - Jing W. Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- CREST, Japan Science and Technology Agency (JST), Gobancho, Chiyoda-ku, Tokyo, Japan
- * E-mail: (TC); (MMi)
| |
Collapse
|
41
|
Roy B, Jackson GR. Interactions between Tau and α-synuclein augment neurotoxicity in a Drosophila model of Parkinson's disease. Hum Mol Genet 2014; 23:3008-23. [PMID: 24430504 DOI: 10.1093/hmg/ddu011] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Clinical and pathological studies have suggested considerable overlap between tauopathies and synucleinopathies. Several genome-wide association studies have identified alpha-Synuclein (SNCA) and Tau (MAPT) polymorphisms as common risk factors for sporadic Parkinson's disease (PD). However, the mechanisms by which subtle variations in the expression of wild-type SNCA and MAPT influence risk for PD and the underlying cellular events that effect neurotoxicity remain unclear. To examine causes of neurotoxicity associated with the α-Syn/Tau interaction, we used the fruit fly as a model. We utilized misexpression paradigms in three different tissues to probe the α-Syn/Tau interaction: the retina, dopaminergic neurons and the larval neuromuscular junction. Misexpression of Tau and α-Syn enhanced a rough eye phenotype and loss of dopaminergic neurons in fly tauopathy and synucleinopathy models, respectively. Our findings suggest that interactions between α-Syn and Tau at the cellular level cause disruption of cytoskeletal organization, axonal transport defects and aberrant synaptic organization that contribute to neuronal dysfunction and death associated with sporadic PD. α-Syn did not alter levels of Tau phosphorylated at the AT8 epitope. However, α-Syn and Tau colocalized in ubiquitin-positive aggregates in eye imaginal discs. The presence of Tau also led to an increase in urea soluble α-Syn. Our findings have important implications in understanding the cellular and molecular mechanisms underlying α-Syn/Tau-mediated synaptic dysfunction, which likely arise in the early asymptomatic phase of sporadic PD.
Collapse
Affiliation(s)
- Bidisha Roy
- Mitchell Center for Neurodegenerative Diseases
| | | |
Collapse
|
42
|
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: 35] [Impact Index Per Article: 3.2] [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
|
43
|
Adult Neurogenesis in Drosophila. Cell Rep 2013; 3:1857-65. [DOI: 10.1016/j.celrep.2013.05.034] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/26/2013] [Accepted: 05/22/2013] [Indexed: 12/28/2022] Open
|
44
|
Abstract
An experimental method has been developed to investigate the cellular responses to central nervous system (CNS) injury using the fruit-fly Drosophila. Understanding repair and regeneration in animals is a key question in biology. The damaged human CNS does not regenerate, and understanding how to promote the regeneration is one of main goals of medical neuroscience. The powerful genetic toolkit of Drosophila can be used to tackle the problem of CNS regeneration. A lesion to the CNS ventral nerve cord (VNC, equivalent to the vertebrate spinal cord) is applied manually with a tungsten needle. The VNC can subsequently be filmed in time-lapse using laser scanning confocal microscopy for up to 24 hr to follow the development of the lesion over time. Alternatively, it can be cultured, then fixed and stained using immunofluorescence to visualize neuron and glial cells with confocal microscopy. Using appropriate markers, changes in cell morphology and cell state as a result of injury can be visualized. With ImageJ and purposely developed plug-ins, quantitative and statistical analyses can be carried out to measure changes in wound size over time and the effects of injury in cell proliferation and cell death. These methods allow the analysis of large sample sizes. They can be combined with the powerful genetics of Drosophila to investigate the molecular mechanisms underlying CNS regeneration and repair.
Collapse
Affiliation(s)
- Kentaro Kato
- Neurodevelopment Group, School of Biosciences, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
45
|
Abstract
Identification and counting of cells is necessary to test biological hypotheses, for instance of nervous system formation, disease, degeneration, injury and regeneration, but manual counting is time consuming, tedious, and subject to bias. The fruit fly Drosophila is a widely used model organism to analyse gene function, and most research is carried out in the intact animal or in whole organs, rather than in cell culture. Inferences on gene function require that cell counts are known from these sample types. Image processing and pattern recognition techniques are appropriate tools to automate cell counting. However, counting cells in Drosophila is a complex task: variations in immunohistochemical markers and developmental stages result in images of very different properties, rendering it challenging to identify true cells. Here, we present a technique for counting automatically larval glial cells in three dimensions, from confocal microscopy serial optical sections. Local outlier thresholding and domes are combined to find the cells. Shape descriptors extracted from a data set are used to characterize cells and avoid oversegmentation. Morphological operators are employed to divide cells that could otherwise be missed. The method is accurate and very fast, and treats all samples equally and objectively, rendering all data comparable across specimens. Our method is also applicable to identify cells labelled with other nuclear markers and in sections of mouse tissues.
Collapse
Affiliation(s)
- Manuel G. Forero
- Neurodevelopment Group, School of Biosciences, University of Birmingham, Birmingham B15 2TT
| | - Kentaro Kato
- Neurodevelopment Group, School of Biosciences, University of Birmingham, Birmingham B15 2TT
| | - Alicia Hidalgo
- Neurodevelopment Group, School of Biosciences, University of Birmingham, Birmingham B15 2TT
| |
Collapse
|
46
|
Read RD. Drosophila melanogaster as a model system for human brain cancers. Glia 2011; 59:1364-76. [PMID: 21538561 PMCID: PMC3221733 DOI: 10.1002/glia.21148] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 12/23/2010] [Indexed: 11/09/2022]
Abstract
Glioblastomas (GBM), the most common primary brain tumors, infiltrate the brain, grow rapidly, and are refractory to current therapies. Signature genetic lesions in glioblastomas include mutation of the epidermal growth factor receptor tyrosine kinase (EGFR) receptor tyrosine kinase and activating mutations in components of the PI-3 kinase (PI3K) pathway. Despite years of study, how these pathways specifically regulate glial pathogenesis is unclear. To address the genetic and cellular origins of this disease, a novel Drosophila GBM model has been developed in which glial progenitor cells give rise to proliferative and invasive neoplastic cells that create transplantable tumors in response to constitutive co-activation of the EGFR-Ras and PI3K pathways. Standing with a rich literature demonstrating the direct relevance of Drosophila to studies on human cancer, neurological disease, and neurodevelopment, this model represents a robust cell-type specific Drosophila neurological disease model in which malignant cells are created by mutations in genetic pathways thought to be driving forces in a homologous human disease. Using lineage analysis and cell-type specific markers, neoplastic glial cells were found to originate from committed glial progenitor cells, rather than from multipotent neuroblasts. Genetic analyses demonstrated that EGFR-Ras and PI3K induce fly glial neoplasia through activation of a combinatorial genetic network composed, in part, of other genetic pathways also commonly mutated in human glioblastomas. In the future, large-scale forward genetic screens with this model may reveal new insights into the origins and treatments of human glioblastoma.
Collapse
Affiliation(s)
- Renee D Read
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA.
| |
Collapse
|
47
|
Kato K, Forero MG, Fenton JC, Hidalgo A. The glial regenerative response to central nervous system injury is enabled by pros-notch and pros-NFκB feedback. PLoS Biol 2011; 9:e1001133. [PMID: 21912512 PMCID: PMC3166069 DOI: 10.1371/journal.pbio.1001133] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 07/21/2011] [Indexed: 01/06/2023] Open
Abstract
Organisms are structurally robust, as cells accommodate changes preserving structural integrity and function. The molecular mechanisms underlying structural robustness and plasticity are poorly understood, but can be investigated by probing how cells respond to injury. Injury to the CNS induces proliferation of enwrapping glia, leading to axonal re-enwrapment and partial functional recovery. This glial regenerative response is found across species, and may reflect a common underlying genetic mechanism. Here, we show that injury to the Drosophila larval CNS induces glial proliferation, and we uncover a gene network controlling this response. It consists of the mutual maintenance between the cell cycle inhibitor Prospero (Pros) and the cell cycle activators Notch and NFκB. Together they maintain glia in the brink of dividing, they enable glial proliferation following injury, and subsequently they exert negative feedback on cell division restoring cell cycle arrest. Pros also promotes glial differentiation, resolving vacuolization, enabling debris clearance and axonal enwrapment. Disruption of this gene network prevents repair and induces tumourigenesis. Using wound area measurements across genotypes and time-lapse recordings we show that when glial proliferation and glial differentiation are abolished, both the size of the glial wound and neuropile vacuolization increase. When glial proliferation and differentiation are enabled, glial wound size decreases and injury-induced apoptosis and vacuolization are prevented. The uncovered gene network promotes regeneration of the glial lesion and neuropile repair. In the unharmed animal, it is most likely a homeostatic mechanism for structural robustness. This gene network may be of relevance to mammalian glia to promote repair upon CNS injury or disease.
Collapse
Affiliation(s)
- Kentaro Kato
- NeuroDevelopment Group, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Manuel G. Forero
- NeuroDevelopment Group, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Janine C. Fenton
- NeuroDevelopment Group, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Alicia Hidalgo
- NeuroDevelopment Group, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
Twohig JP, Cuff SM, Yong AA, Wang ECY. The role of tumor necrosis factor receptor superfamily members in mammalian brain development, function and homeostasis. Rev Neurosci 2011; 22:509-33. [PMID: 21861782 DOI: 10.1515/rns.2011.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor receptor superfamily (TNFRSF) members were initially identified as immunological mediators, and are still commonly perceived as immunological molecules. However, our understanding of the diversity of TNFRSF members' roles in mammalian physiology has grown significantly since the first discovery of TNFRp55 (TNFRSF1) in 1975. In particular, the last decade has provided evidence for important roles in brain development, function and the emergent field of neuronal homeostasis. Recent evidence suggests that TNFRSF members are expressed in an overlapping regulated pattern during neuronal development, participating in the regulation of neuronal expansion, growth, differentiation and regional pattern development. This review examines evidence for non-immunological roles of TNFRSF members in brain development, function and maintenance under normal physiological conditions. In addition, several aspects of brain function during inflammation will also be described, when illuminating and relevant to the non-immunological role of TNFRSF members. Finally, key questions in the field will be outlined.
Collapse
Affiliation(s)
- Jason P Twohig
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, Wales, UK
| | | | | | | |
Collapse
|
49
|
Gatto CL, Broadie K. Fragile X mental retardation protein is required for programmed cell death and clearance of developmentally-transient peptidergic neurons. Dev Biol 2011; 356:291-307. [PMID: 21596027 PMCID: PMC3143227 DOI: 10.1016/j.ydbio.2011.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 04/22/2011] [Accepted: 05/03/2011] [Indexed: 01/19/2023]
Abstract
Fragile X syndrome (FXS), caused by loss of fragile X mental retardation 1 (FMR1) gene function, is the most common heritable cause of intellectual disability and autism spectrum disorders. The FMR1 product (FMRP) is an RNA-binding protein best established to function in activity-dependent modulation of synaptic connections. In the Drosophila FXS disease model, loss of functionally-conserved dFMRP causes synaptic overgrowth and overelaboration in pigment dispersing factor (PDF) peptidergic neurons in the adult brain. Here, we identify a very different component of PDF neuron misregulation in dfmr1 mutants: the aberrant retention of normally developmentally-transient PDF tritocerebral (PDF-TRI) neurons. In wild-type animals, PDF-TRI neurons in the central brain undergo programmed cell death and complete, processive clearance within days of eclosion. In the absence of dFMRP, a defective apoptotic program leads to constitutive maintenance of these peptidergic neurons. We tested whether this apoptotic defect is circuit-specific by examining crustacean cardioactive peptide (CCAP) and bursicon circuits, which are similarly developmentally-transient and normally eliminated immediately post-eclosion. In dfmr1 null mutants, CCAP/bursicon neurons also exhibit significantly delayed clearance dynamics, but are subsequently eliminated from the nervous system, in contrast to the fully persistent PDF-TRI neurons. Thus, the requirement of dFMRP for the retention of transitory peptidergic neurons shows evident circuit specificity. The novel defect of impaired apoptosis and aberrant neuron persistence in the Drosophila FXS model suggests an entirely new level of "pruning" dysfunction may contribute to the FXS disease state.
Collapse
Affiliation(s)
- Cheryl L Gatto
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
50
|
Neural Stem Cell Biology in Vertebrates and Invertebrates: More Alike than Different? Neuron 2011; 70:719-29. [DOI: 10.1016/j.neuron.2011.05.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2011] [Indexed: 11/20/2022]
|