1
|
Kwon SY, Chan K, Stofanko M, Chan KH, Badenhorst P. Abrupt-mediated control of ninjurins regulates Drosophila sessile haemocyte compartments. Development 2024; 151:dev202977. [PMID: 39545919 DOI: 10.1242/dev.202977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Macrophage-like cells called haemocytes are key effectors of Drosophila cellular innate immune function. Larval haemocytes exist either in circulation or localize to segmentally repeated sessile haemocyte compartments (SHCs). While numerous functions have been proposed for SHCs, the mechanisms directing haemocytes to them are unclear. Here, we have exploited the developmentally regulated dispersal of SHCs that occurs at pupariation to identify the Abrupt (Ab) transcription factor (TF) and ninjurin cell-adhesion molecules as regulators of haemocyte recruitment to SHCs. We show that larval haemocytes express ninjurins, which are required for targeting haemocytes to SHCs. However, at pupariation, ecdysteroid signalling stimulates Ab expression, which collaborates with TFs, including Blimp-1 and Hr3, to repress ninjurins and disperse haemocytes. We observe that experimental manipulations that antagonize ninjurin function in larval haemocytes cause premature SHC dispersal, while stabilization of ninjurins in haemocytes blocks developmentally regulated SHC remodelling and increases sensitivity to immune challenges. Cumulatively, our data indicate that control of ninjurin activity provides a common target through which diverse developmental, environmental and immune stimuli can be integrated to control haemocyte dispersal and immune function.
Collapse
Affiliation(s)
- So Yeon Kwon
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Kimberly Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Martin Stofanko
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Ka Hei Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Paul Badenhorst
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
2
|
Deichsel S, Frankenreiter L, Fechner J, Gahr BM, Zimmermann M, Mastel H, Preis I, Preiss A, Nagel AC. Inhibition of the Notch signal transducer CSL by Pkc53E-mediated phosphorylation to fend off parasitic immune challenge in Drosophila. eLife 2024; 12:RP89582. [PMID: 39503739 PMCID: PMC11540305 DOI: 10.7554/elife.89582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Notch signalling activity regulates hematopoiesis in Drosophila and vertebrates alike. Parasitoid wasp infestation of Drosophila larvae, however, requires a timely downregulation of Notch activity to allow the formation of encapsulation-active blood cells. Here, we show that the Drosophila CSL transcription factor Suppressor of Hairless [Su(H)] is phosphorylated at Serine 269 in response to parasitoid wasp infestation. As this phosphorylation interferes with the DNA binding of Su(H), it reversibly precludes its activity. Accordingly, phospho-deficient Su(H)S269A mutants are immune-compromised. A screen for kinases involved in Su(H) phosphorylation identified Pkc53E, required for normal hematopoiesis as well as for parasitoid immune response. Genetic and molecular interactions support the specificity of the Su(H)-Pkc53E relationship. Moreover, phorbol ester treatment inhibits Su(H) activity in vivo and in human cell culture. We conclude that Pkc53E targets Su(H) during parasitic wasp infestation, thereby remodelling the blood cell population required for wasp egg encapsulation.
Collapse
Affiliation(s)
- Sebastian Deichsel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Medical Genetics and Applied Genomics, University of TübingenTübingenGermany
| | - Lisa Frankenreiter
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Johannes Fechner
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Institute of Biomedical Genetics (IBMG), University of StuttgartStuttgartGermany
| | - Bernd M Gahr
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
- Department of Internal Medicine II, Molecular Cardiology, University of UlmUlmGermany
| | - Mirjam Zimmermann
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Helena Mastel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Irina Preis
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anette Preiss
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| | - Anja C Nagel
- Department of Molecular Genetics, Institute of Biology, University of HohenheimStuttgartGermany
| |
Collapse
|
3
|
Chechenova M, McLendon L, Dallas B, Stratton H, Kiani K, Gerberich E, Alekseyenko A, Tamba N, An S, Castillo L, Czajkowski E, Talley C, Brown A, Bryantsev AL. Muscle degeneration in aging Drosophila flies: the role of mechanical stress. Skelet Muscle 2024; 14:20. [PMID: 39164781 PMCID: PMC11334408 DOI: 10.1186/s13395-024-00352-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
Muscle wasting is a universal hallmark of aging which is displayed by a wide range of organisms, although the causes and mechanisms of this phenomenon are not fully understood. We used Drosophila to characterize the phenomenon of spontaneous muscle fiber degeneration (SMFD) during aging. We found that SMFD occurs across diverse types of somatic muscles, progresses with chronological age, and positively correlates with functional muscle decline. Data from vital dyes and morphological markers imply that degenerative fibers most likely die by necrosis. Mechanistically, SMFD is driven by the damage resulting from muscle contractions, and the nervous system may play a significant role in this process. Our quantitative model of SMFD assessment can be useful in identifying and validating novel genetic factors that influence aging-related muscle wasting.
Collapse
Affiliation(s)
- Maria Chechenova
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
- Present Affiliation: MNG Laboratories, A LabCorp Company, Atlanta, GA, USA
| | - Lilla McLendon
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Bracey Dallas
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Hannah Stratton
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Kaveh Kiani
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Erik Gerberich
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Alesia Alekseyenko
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Natasya Tamba
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - SooBin An
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Lizzet Castillo
- Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Emily Czajkowski
- Department of Biology, University of New Mexico, Albuquerque, NM, USA
- Present Affiliation: Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Christina Talley
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA
| | - Austin Brown
- Department of Mathematics, Kennesaw State University, Kennesaw, GA, USA
| | - Anton L Bryantsev
- Department of Molecular and Cellular Biology, Kennesaw State University, 105 Marietta Dr., NW, Room 4004, MD 1201, Kennesaw, GA, 30144, USA.
| |
Collapse
|
4
|
Doyle EH, Vaughan HJ, Mariani SA. From drosophila to humans: a journey through macrophage development. Exp Hematol 2024; 136:104272. [PMID: 38972565 DOI: 10.1016/j.exphem.2024.104272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Macrophages are fascinating immune cells involved in a variety of processes in both health and disease. Although they were first discovered and characterized by their functions as professional phagocytes and antigen-presenting cells, it is now clear that macrophages have multiple roles within embryonic development, tissue homeostasis, regulation of inflammation, and host response to pathogens and tissue insults. Interestingly, macrophages, or macrophage-like cells, exist in a variety of organisms, from echinoderms to humans, and are present also in species that lack an adaptive immune system or hematopoietic stem cells (HSCs). In mammals, macrophages can be generated from bone marrow precursors through a monocyte intermediate, but it is now known that they are also generated during earlier hematopoietic waves in the embryo. Seeding a variety of tissues at different times, macrophages contribute to embryonic organogenesis and tissue homeostasis. Interestingly, in species where embryonic macrophages are generated before HSC specification, they seem to be an important component of the HSC generative microenvironment. There are many excellent reviews reporting the current knowledge on the ontogeny and functions of macrophages in adult tissues. Here, we aim to summarize the current knowledge on the development and functions of embryonic macrophages across the most used animal models, with a special focus on developmental hematopoiesis.
Collapse
Affiliation(s)
- Eva H Doyle
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hollie J Vaughan
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom
| | - Samanta A Mariani
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
5
|
Fioriti F, Rifflet A, Gomperts Boneca I, Zugasti O, Royet J. Bacterial peptidoglycan serves as a critical modulator of the gut-immune-brain axis in Drosophila. Brain Behav Immun 2024; 119:878-897. [PMID: 38710338 DOI: 10.1016/j.bbi.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
Metabolites and compounds derived from gut-associated bacteria can modulate numerous physiological processes in the host, including immunity and behavior. Using a model of oral bacterial infection, we previously demonstrated that gut-derived peptidoglycan (PGN), an essential constituent of the bacterial cell envelope, influences female fruit fly egg-laying behavior by activating the NF-κB cascade in a subset of brain neurons. These findings underscore PGN as a potential mediator of communication between gut bacteria and the brain in Drosophila, prompting further investigation into its impact on all brain cells. Through high-resolution mass spectrometry, we now show that PGN fragments produced by gut bacteria can rapidly reach the central nervous system. In Addition, by employing a combination of whole-genome transcriptome analyses, comprehensive genetic assays, and reporter gene systems, we reveal that gut bacterial infection triggers a PGN dose-dependent NF-κB immune response in perineurial glia, forming the continuous outer cell layer of the blood-brain barrier. Furthermore, we demonstrate that persistent PGN-dependent NF-κB activation in perineurial glial cells correlates with a reduction in lifespan and early neurological decline. Overall, our findings establish gut-derived PGN as a critical mediator of the gut-immune-brain axis in Drosophila.
Collapse
Affiliation(s)
- Florent Fioriti
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France
| | - Aline Rifflet
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, 75015 Paris, France
| | - Ivo Gomperts Boneca
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, 75015 Paris, France
| | - Olivier Zugasti
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France.
| | - Julien Royet
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France.
| |
Collapse
|
6
|
Hersperger F, Kastl M, Paeschke K, Kierdorf K. Hemocyte Nuclei Isolation from Adult Drosophila melanogaster for snRNA-seq. Methods Mol Biol 2024; 2713:71-79. [PMID: 37639115 DOI: 10.1007/978-1-0716-3437-0_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
In adult Drosophila, most of the hemocytes are macrophage-like cells (so called plasmatocytes), which serve various functions in organ homeostasis and immune defense. Ontogeny and functions are largely conserved between vertebrate and invertebrate macrophages. Hence, Drosophila offers a powerful genetic toolbox to study macrophage function and genetically modulate these cells. Technological advances in high-throughput sequencing approaches allowed to give an in-depth characterization of vertebrate macrophage populations and their heterogenous composition within different organs as well as changes in disease. Embryonic and larval hemocytes in Drosophila have been recently analyzed in single-cell RNA-sequencing (scRNA-seq) approaches during infection and steady state. These analyses revealed anatomical and functional Drosophila hemocyte subtypes dedicated to specific tasks. Only recently, the Fly Cell Atlas provided a whole transcriptomic single-cell atlas via single-nuclei RNA-sequencing (snRNA-seq) of adult Drosophila including many different tissues and cell types where hemocytes were also included. Yet, a specific protocol to isolate nuclei from adult hemocytes for snRNA-seq and study these cells in different experimental conditions was not available. In this chapter, we give a detailed protocol to purify hemocyte nuclei from adult Drosophila, which can be used in subsequent analyses such as snRNA-seq.
Collapse
Affiliation(s)
- Fabian Hersperger
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Kastl
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Katrin Paeschke
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany.
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
7
|
Hastings CJ, Keledjian MV, Musselman LP, Marques CNH. Delayed host mortality and immune response upon infection with P. aeruginosa persister cells. Infect Immun 2023; 91:e0024623. [PMID: 37732789 PMCID: PMC10580972 DOI: 10.1128/iai.00246-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 09/22/2023] Open
Abstract
Chronic infections are a heavy burden on healthcare systems worldwide. Persister cells are thought to be largely responsible for chronic infection due to their tolerance to antimicrobials and recalcitrance to innate immunity factors. Pseudomonas aeruginosa is a common and clinically relevant pathogen that contains stereotypical persister cells. Despite their importance in chronic infection, there have been limited efforts to study persister cell infections in vivo. Drosophila melanogaster has a well-described innate immune response similar to that of vertebrates and is a good candidate for the development of an in vivo model of infection for persister cells. Similar to what is observed in other bacterial strains, in this work we found that infection with P. aeruginosa persister cells resulted in a delayed mortality phenotype in Caenorhabditis elegans, Arabidopsis thaliana, and D. melanogaster compared to infection with regular cells. An in-depth characterization of infected D. melanogaster found that bacterial loads differed between persister and regular cells' infections during the early stages. Furthermore, hemocyte activation and antimicrobial peptide expression were delayed/reduced in persister infections over the same time course, indicating an initial suppression of, or inability to elicit, the fly immune response. Overall, our findings support the use of D. melanogaster as a model in which to study persister cells in vivo, where this bacterial subpopulation exhibits delayed virulence and an attenuated immune response.
Collapse
Affiliation(s)
- Cody J. Hastings
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | - Maya V. Keledjian
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | | | - Cláudia N. H. Marques
- Department of Biological Sciences, Binghamton University, Binghamton, New York, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
8
|
Hirschhäuser A, Molitor D, Salinas G, Großhans J, Rust K, Bogdan S. Single-cell transcriptomics identifies new blood cell populations in Drosophila released at the onset of metamorphosis. Development 2023; 150:dev201767. [PMID: 37681301 PMCID: PMC10560556 DOI: 10.1242/dev.201767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
Drosophila blood cells called hemocytes form an efficient barrier against infections and tissue damage. During metamorphosis, hemocytes undergo tremendous changes in their shape and behavior, preparing them for tissue clearance. Yet, the diversity and functional plasticity of pupal blood cells have not been explored. Here, we combine single-cell transcriptomics and high-resolution microscopy to dissect the heterogeneity and plasticity of pupal hemocytes. We identified undifferentiated and specified hemocytes with different molecular signatures associated with distinct functions such as antimicrobial, antifungal immune defense, cell adhesion or secretion. Strikingly, we identified a highly migratory and immune-responsive pupal cell population expressing typical markers of the posterior signaling center (PSC), which is known to be an important niche in the larval lymph gland. PSC-like cells become restricted to the abdominal segments and are morphologically very distinct from typical Hemolectin (Hml)-positive plasmatocytes. G-TRACE lineage experiments further suggest that PSC-like cells can transdifferentiate to lamellocytes triggered by parasitoid wasp infestation. In summary, we present the first molecular description of pupal Drosophila blood cells, providing insights into blood cell functional diversification and plasticity during pupal metamorphosis.
Collapse
Affiliation(s)
- Alexander Hirschhäuser
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Darius Molitor
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University Marburg, Karl-von-Frisch-Strasse 8, 35043 Marburg, Germany
| | - Katja Rust
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| |
Collapse
|
9
|
Voutyraki C, Choromidis A, Meligkounaki A, Vlachopoulos NA, Theodorou V, Grammenoudi S, Athanasiadis E, Monticelli S, Giangrande A, Delidakis C, Zacharioudaki E. Growth deregulation and interaction with host hemocytes contribute to tumor progression in a Drosophila brain tumor model. Proc Natl Acad Sci U S A 2023; 120:e2221601120. [PMID: 37549261 PMCID: PMC10438840 DOI: 10.1073/pnas.2221601120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 08/09/2023] Open
Abstract
Tumors constantly interact with their microenvironment. Here, we present data on a Notch-induced neural stem cell (NSC) tumor in Drosophila, which can be immortalized by serial transplantation in adult hosts. This tumor arises in the larva by virtue of the ability of Notch to suppress early differentiation-promoting factors in NSC progeny. Guided by transcriptome data, we have addressed both tumor-intrinsic and microenvironment-specific factors and how they contribute to tumor growth and host demise. The growth promoting factors Myc, Imp, and Insulin receptor in the tumor cells are important for tumor expansion and killing of the host. From the host's side, hemocytes, professional phagocytic blood cells, are found associated with tumor cells. Phagocytic receptors, like NimC1, are needed in hemocytes to enable them to capture and engulf tumor cells, restricting their growth. In addition to their protective role, hemocytes may also increase the host's morbidity by their propensity to produce damaging extracellular reactive oxygen species.
Collapse
Affiliation(s)
- Chrysanthi Voutyraki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Alexandros Choromidis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Anastasia Meligkounaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Nikolaos Andreas Vlachopoulos
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Vasiliki Theodorou
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| | - Sofia Grammenoudi
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center Alexander Fleming, 16672Athens, Greece
| | - Emmanouil Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, 11527Athens, Greece
- Medical Image and Signal Processing Laboratory, Department of Biomedical Engineering, University of West Attica, 12243Athens, Greece
| | - Sara Monticelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400Strasbourg, France
- Centre National de la Recherche Scientifique, UMR7104Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1258Strasbourg, France
- Université de Strasbourg, 67404Strasbourg, France
| | - Christos Delidakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
- Department of Biology, University of Crete, 70013Heraklion, Crete, Greece
| | - Evanthia Zacharioudaki
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, 70013Heraklion, Crete, Greece
| |
Collapse
|
10
|
Abstract
Endocrine signaling networks control diverse biological processes and life history traits across metazoans. In both invertebrate and vertebrate taxa, steroid hormones regulate immune system function in response to intrinsic and environmental stimuli, such as microbial infection. The mechanisms of this endocrine-immune regulation are complex and constitute an ongoing research endeavor facilitated by genetically tractable animal models. The 20-hydroxyecdysone (20E) is the major steroid hormone in arthropods, primarily studied for its essential role in mediating developmental transitions and metamorphosis; 20E also modulates innate immunity in a variety of insect taxa. This review provides an overview of our current understanding of 20E-mediated innate immune responses. The prevalence of correlations between 20E-driven developmental transitions and innate immune activation are summarized across a range of holometabolous insects. Subsequent discussion focuses on studies conducted using the extensive genetic resources available in Drosophila that have begun to reveal the mechanisms underlying 20E regulation of immunity in the contexts of both development and bacterial infection. Lastly, I propose directions for future research into 20E regulation of immunity that will advance our knowledge of how interactive endocrine networks coordinate animals' physiological responses to environmental microbes.
Collapse
Affiliation(s)
- Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
11
|
Bossen J, Kühle JP, Roeder T. The tracheal immune system of insects - A blueprint for understanding epithelial immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103960. [PMID: 37235953 DOI: 10.1016/j.ibmb.2023.103960] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
The unique design of respiratory organs in multicellular organisms makes them prone to infection by pathogens. To cope with this vulnerability, highly effective local immune systems evolved that are also operative in the tracheal system of insects. Many pathogens and parasites (including viruses, bacteria, fungi, and metazoan parasites) colonize the trachea or invade the host via this route. Currently, only two modules of the tracheal immune system have been characterized in depth: 1) Immune deficiency pathway-mediated activation of antimicrobial peptide gene expression and 2) local melanization processes that protect the structure from wounding. There is an urgent need to increase our understanding of the architecture of tracheal immune systems, especially regarding those mechanisms that enable the maintenance of immune homeostasis. This need for new studies is particularly exigent for species other than Drosophila.
Collapse
Affiliation(s)
- Judith Bossen
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Jan-Philip Kühle
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany
| | - Thomas Roeder
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
12
|
Touré H, Galindo LA, Lagune M, Glatigny S, Waterhouse RM, Guénal I, Herrmann JL, Girard-Misguich F, Szuplewski S. Mycobacterium abscessus resists the innate cellular response by surviving cell lysis of infected phagocytes. PLoS Pathog 2023; 19:e1011257. [PMID: 36972320 PMCID: PMC10079227 DOI: 10.1371/journal.ppat.1011257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/06/2023] [Accepted: 02/28/2023] [Indexed: 03/29/2023] Open
Abstract
Mycobacterium abscessus is the most pathogenic species among the predominantly saprophytic fast-growing mycobacteria. This opportunistic human pathogen causes severe infections that are difficult to eradicate. Its ability to survive within the host was described mainly with the rough (R) form of M. abscessus, which is lethal in several animal models. This R form is not present at the very beginning of the disease but appears during the progression and the exacerbation of the mycobacterial infection, by transition from a smooth (S) form. However, we do not know how the S form of M. abscessus colonizes and infects the host to then multiply and cause the disease. In this work, we were able to show the hypersensitivity of fruit flies, Drosophila melanogaster, to intrathoracic infections by the S and R forms of M. abscessus. This allowed us to unravel how the S form resists the innate immune response developed by the fly, both the antimicrobial peptides- and cellular-dependent immune responses. We demonstrate that intracellular M. abscessus was not killed within the infected phagocytic cells, by resisting lysis and caspase-dependent apoptotic cell death of Drosophila infected phagocytes. In mice, in a similar manner, intra-macrophage M. abscessus was not killed when M. abscessus-infected macrophages were lysed by autologous natural killer cells. These results demonstrate the propensity of the S form of M. abscessus to resist the host’s innate responses to colonize and multiply within the host.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Lee Ann Galindo
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Marion Lagune
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Simon Glatigny
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
| | - Robert M. Waterhouse
- Department of Ecology and Evolution, University of Lausanne, and the Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Ile-de-France Ouest, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-Le-Bretonneux, France
- * E-mail: (FGM); (SS)
| | | |
Collapse
|
13
|
Bossen J, Prange R, Kühle JP, Künzel S, Niu X, Hammel JU, Krieger L, Knop M, Ehrhardt B, Uliczka K, Krauss-Etschmann S, Roeder T. Adult and Larval Tracheal Systems Exhibit Different Molecular Architectures in Drosophila. Int J Mol Sci 2023; 24:ijms24065628. [PMID: 36982710 PMCID: PMC10052349 DOI: 10.3390/ijms24065628] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Knowing the molecular makeup of an organ system is required for its in-depth understanding. We analyzed the molecular repertoire of the adult tracheal system of the fruit fly Drosophila melanogaster using transcriptome studies to advance our knowledge of the adult insect tracheal system. Comparing this to the larval tracheal system revealed several major differences that likely influence organ function. During the transition from larval to adult tracheal system, a shift in the expression of genes responsible for the formation of cuticular structure occurs. This change in transcript composition manifests in the physical properties of cuticular structures of the adult trachea. Enhanced tonic activation of the immune system is observed in the adult trachea, which encompasses the increased expression of antimicrobial peptides. In addition, modulatory processes are conspicuous, in this case mainly by the increased expression of G protein-coupled receptors in the adult trachea. Finally, all components of a peripheral circadian clock are present in the adult tracheal system, which is not the case in the larval tracheal system. Comparative analysis of driver lines targeting the adult tracheal system revealed that even the canonical tracheal driver line breathless (btl)-Gal4 is not able to target all parts of the adult tracheal system. Here, we have uncovered a specific transcriptome pattern of the adult tracheal system and provide this dataset as a basis for further analyses of the adult insect tracheal system.
Collapse
Affiliation(s)
- Judith Bossen
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
| | - Ruben Prange
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Jan-Philip Kühle
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Sven Künzel
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, 24306 Plön, Germany
| | - Xiao Niu
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Jörg U. Hammel
- Helmholtz-Zentrum Hereon, Institute of Materials Physics, 21502 Geesthacht, Germany
| | - Laura Krieger
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Mirjam Knop
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Birte Ehrhardt
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
| | - Karin Uliczka
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
| | - Susanne Krauss-Etschmann
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
- Research Center Borstel, Priority Research Area Chronic Lung Diseases, Early Life Origins of CLD, 23485 Borstel, Germany
- Institute for Experimental Medicine, Kiel University, 24118 Kiel, Germany
| | - Thomas Roeder
- Department Zoology, Molecular Physiology, Kiel University, 24118 Kiel, Germany
- German Lung Center (DZL), Airway Research Center North (ARCN), 24118 Kiel, Germany
- Correspondence: ; Tel.: +49-431-880-81
| |
Collapse
|
14
|
Chechenova M, Stratton H, Kiani K, Gerberich E, Alekseyenko A, Tamba N, An S, Castillo L, Czajkowski E, Talley C, Bryantsev A. Quantitative model of aging-related muscle degeneration: a Drosophila study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.19.529145. [PMID: 36865342 PMCID: PMC9980004 DOI: 10.1101/2023.02.19.529145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Changes in the composition and functionality of somatic muscles is a universal hallmark of aging that is displayed by a wide range of species. In humans, complications arising from muscle decline due to sarcopenia aggravate morbidity and mortality rates. The genetics of aging-related deterioration of muscle tissue is not well understood, which prompted us to characterize aging-related muscle degeneration in Drosophila melanogaster (fruit fly), a leading model organism in experimental genetics. Adult flies demonstrate spontaneous degeneration of muscle fibers in all types of somatic muscles, which correlates with functional, chronological, and populational aging. Morphological data imply that individual muscle fibers die by necrosis. Using quantitative analysis, we demonstrate that muscle degeneration in aging flies has a genetic component. Chronic neuronal overstimulation of muscles promotes fiber degeneration rates, suggesting a role for the nervous system in muscle aging. From the other hand, muscles decoupled from neuronal stimulation retain a basal level of spontaneous degeneration, suggesting the presence of intrinsic factors. Based on our characterization, Drosophila can be adopted for systematic screening and validation of genetic factors linked to aging-related muscle loss.
Collapse
Affiliation(s)
- Maria Chechenova
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Hannah Stratton
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Kaveh Kiani
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Erik Gerberich
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Alesia Alekseyenko
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Natasya Tamba
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - SooBin An
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Lizzet Castillo
- Department of Biology, University of New Mexico, Albuquerque, NM
| | - Emily Czajkowski
- Department of Biology, University of New Mexico, Albuquerque, NM
| | - Christina Talley
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| | - Anton Bryantsev
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA
| |
Collapse
|
15
|
Giordani G, Cattabriga G, Becchimanzi A, Di Lelio I, De Leva G, Gigliotti S, Pennacchio F, Gargiulo G, Cavaliere V. Role of neuronal and non-neuronal acetylcholine signaling in Drosophila humoral immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 153:103899. [PMID: 36596348 DOI: 10.1016/j.ibmb.2022.103899] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Acetylcholine (ACh) is one the major neurotransmitters in insects, whose role in mediating synaptic interactions between neurons in the central nervous system is well characterized. It also plays largely unexplored regulatory functions in non-neuronal tissues. Here we demonstrate that ACh signaling is involved in the modulation of the innate immune response of Drosophila melanogaster. Knockdown of ACh synthesis or ACh vesicular transport in neurons reduced the activation of drosomycin (drs), a gene encoding an antimicrobial peptide, in adult flies infected with a Gram-positive bacterium. drs transcription was similarly affected in Drosophila α7 nicotinic acetylcholine receptor, nAChRalpha7 (Dα7) mutants, as well as in flies expressing in the nervous system a dominant negative form (Dα7DN) of this specific receptor subunit. Interestingly, Dα7DN elicited a comparable response when it was expressed in non-neuronal tissues and even when it was specifically produced in the hemocytes. Consistently, full activation of the drs gene required Dα7 expression in these cells. Moreover, knockdown of ACh synthesis in non-neuronal cells affected drs expression. Overall, these findings uncover neural and non-neural cholinergic signals that modulate insect immune defenses and shed light on the role of hemocytes in the regulation of the humoral immune response.
Collapse
Affiliation(s)
- Giorgia Giordani
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Giulia Cattabriga
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Andrea Becchimanzi
- Dipartimento di Agraria, Laboratorio di Entomologia "E. Tremblay", Università degli Studi di Napoli "Federico II", Portici, Napoli, Italy
| | - Ilaria Di Lelio
- Dipartimento di Agraria, Laboratorio di Entomologia "E. Tremblay", Università degli Studi di Napoli "Federico II", Portici, Napoli, Italy
| | - Giovanna De Leva
- Dipartimento di Agraria, Laboratorio di Entomologia "E. Tremblay", Università degli Studi di Napoli "Federico II", Portici, Napoli, Italy
| | - Silvia Gigliotti
- Istituto di Bioscienze e Biorisorse, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Francesco Pennacchio
- Dipartimento di Agraria, Laboratorio di Entomologia "E. Tremblay", Università degli Studi di Napoli "Federico II", Portici, Napoli, Italy; Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BATCenter), University of Napoli "Federico II", Portici, NA, Italy.
| | - Giuseppe Gargiulo
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy; Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BATCenter), University of Napoli "Federico II", Portici, NA, Italy.
| | - Valeria Cavaliere
- Dipartimento di Farmacia e Biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italy; Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BATCenter), University of Napoli "Federico II", Portici, NA, Italy.
| |
Collapse
|
16
|
Vaibhvi V, Künzel S, Roeder T. Hemocytes and fat body cells, the only professional immune cell types in Drosophila, show strikingly different responses to systemic infections. Front Immunol 2022; 13:1040510. [PMID: 36505446 PMCID: PMC9726733 DOI: 10.3389/fimmu.2022.1040510] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
The fruit fly Drosophila is an excellent model to study the response of different immunocompetent organs during systemic infection. In the present study, we intended to test the hypothesis that the only professional immune organs of the fly, the fat body and hemocytes, show substantial similarities in their responses to systemic infection. However, comprehensive transcriptome analysis of isolated organs revealed highly divergent transcript signatures, with the few commonly regulated genes encoding mainly classical immune effectors from the antimicrobial peptide family. The fat body and the hemocytes each have specific reactions that are not present in the other organ. Fat body-specific responses comprised those enabling an improved peptide synthesis and export. This reaction is accompanied by transcriptomic shifts enabling the use of the energy resources of the fat body more efficiently. Hemocytes, on the other hand, showed enhanced signatures related to phagocytosis. Comparing immune-induced signatures of both cell types with those of whole-body responses showed only a minimal correspondence, mostly restricted again to antimicrobial peptide genes. In summary, the two major immunocompetent cell types of Drosophila show highly specific responses to infection, which are closely linked to the primary function of the respective organ in the landscape of the systemic immune response.
Collapse
Affiliation(s)
- Vaibhvi Vaibhvi
- Department of Molecular Physiology, Zoology Institute, Kiel University, Kiel, Germany
| | - Sven Künzel
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Thomas Roeder
- Department of Molecular Physiology, Zoology Institute, Kiel University, Kiel, Germany,German Center for Lung Research, Airway Research Center North, Kiel, Germany,*Correspondence: Thomas Roeder,
| |
Collapse
|
17
|
Kinoshita S, Takarada K, Kinoshita Y, Inoue YH. Drosophila hemocytes recognize lymph gland tumors of mxc mutants and activate the innate immune pathway in a reactive oxygen species-dependent manner. Biol Open 2022; 11:bio059523. [PMID: 36226812 PMCID: PMC9641529 DOI: 10.1242/bio.059523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 12/29/2022] Open
Abstract
Mechanisms of cancer cell recognition and elimination by the innate immune system remains unclear. The immune signaling pathways are activated in the fat body to suppress the tumor growth in mxcmbn1 hematopoietic tumor mutants in Drosophila by inducing antimicrobial peptides (AMP). Here, we investigated the regulatory mechanism underlying the activation in the mutant. Firstly, we found that reactive oxygen species (ROS) accumulated in the hemocytes due to induction of dual oxidase and one of its activators. This was required for the AMP induction and the tumor growth suppression. Next, more hemocytes transplanted from normal larvae were associated with the mutant tumor than normal lymph glands (LGs). Matrix metalloproteinase 1 and 2 (MMP2) were highly expressed in the tumors. The basement membrane components in the tumors were reduced and ultimately lost inside. Depletion of the MMP2 rather than MMP1 resulted in a significantly reduced AMP expression in the mutant larvae. The hemocytes may recognize the disassembly of basement membrane in the tumors and activate the ROS production. Our findings highlight the mechanism via which macrophage-like hemocytes recognize tumor cells and subsequently convey the information to induce AMPs in the fat body. They contribute to uncover the role of innate immune system against cancer.
Collapse
Affiliation(s)
- Suzuko Kinoshita
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Kazuki Takarada
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yuriko Kinoshita
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| | - Yoshihiro H. Inoue
- Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto, 606-8585, Japan
| |
Collapse
|
18
|
Fuse N, Okamori C, Okaji R, Tang C, Hirai K, Kurata S. Transcriptome features of innate immune memory in Drosophila. PLoS Genet 2022; 18:e1010005. [PMID: 36252180 PMCID: PMC9612818 DOI: 10.1371/journal.pgen.1010005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 10/27/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022] Open
Abstract
Immune memory is the ability of organisms to elicit potentiated immune responses at secondary infection. Current studies have revealed that similar to adaptive immunity, innate immunity exhibits memory characteristics (called "innate immune memory"). Although epigenetic reprogramming plays an important role in innate immune memory, the underlying mechanisms have not been elucidated, especially at the individual level. Here, we established experimental systems for detecting innate immune memory in Drosophila melanogaster. Training infection with low-pathogenic bacteria enhanced the survival rate of the flies at subsequent challenge infection with high-pathogenic bacteria. Among low-pathogenic bacteria, Micrococcus luteus (Ml) and Salmonella typhimurium (St) exerted apparent training effects in the fly but exhibited different mechanisms of action. Ml exerted training effects even after its clearance from flies, while live St persisted in the flies for a prolonged duration. RNA sequencing (RNA-Seq) analysis revealed that Ml training enhanced the expression of the immune-related genes under the challenge condition but not under the non-challenge condition. In contrast, St training upregulated the expression of the immune-related genes independent of challenge. These results suggest that training effects with Ml and St are due to memory and persistence of immune responses, respectively. Furthermore, we searched for the gene involved in immune memory, and identified a candidate gene, Ada2b, which encodes a component of the histone modification complex. The Ada2b mutant suppressed Ml training effects on survival and disrupted the expression of some genes under the training + challenge condition. These results suggest that the gene expression regulated by Ada2b may contribute to innate immune memory in Drosophila.
Collapse
Affiliation(s)
- Naoyuki Fuse
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail: (NF); (SK)
| | - Chisaki Okamori
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoma Okaji
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Chang Tang
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kikuko Hirai
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- * E-mail: (NF); (SK)
| |
Collapse
|
19
|
Corbally MK, Regan JC. Fly immunity comes of age: The utility of Drosophila as a model for studying variation in immunosenescence. FRONTIERS IN AGING 2022; 3:1016962. [PMID: 36268532 PMCID: PMC9576847 DOI: 10.3389/fragi.2022.1016962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Affiliation(s)
| | - Jennifer C. Regan
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Hultmark D, Andó I. Hematopoietic plasticity mapped in Drosophila and other insects. eLife 2022; 11:e78906. [PMID: 35920811 PMCID: PMC9348853 DOI: 10.7554/elife.78906] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/20/2022] [Indexed: 12/12/2022] Open
Abstract
Hemocytes, similar to vertebrate blood cells, play important roles in insect development and immunity, but it is not well understood how they perform their tasks. New technology, in particular single-cell transcriptomic analysis in combination with Drosophila genetics, may now change this picture. This review aims to make sense of recently published data, focusing on Drosophila melanogaster and comparing to data from other drosophilids, the malaria mosquito, Anopheles gambiae, and the silkworm, Bombyx mori. Basically, the new data support the presence of a few major classes of hemocytes: (1) a highly heterogenous and plastic class of professional phagocytes with many functions, called plasmatocytes in Drosophila and granular cells in other insects. (2) A conserved class of cells that control melanin deposition around parasites and wounds, called crystal cells in D. melanogaster, and oenocytoids in other insects. (3) A new class of cells, the primocytes, so far only identified in D. melanogaster. They are related to cells of the so-called posterior signaling center of the larval hematopoietic organ, which controls the hematopoiesis of other hemocytes. (4) Different kinds of specialized cells, like the lamellocytes in D. melanogaster, for the encapsulation of parasites. These cells undergo rapid evolution, and the homology relationships between such cells in different insects are uncertain. Lists of genes expressed in the different hemocyte classes now provide a solid ground for further investigation of function.
Collapse
Affiliation(s)
- Dan Hultmark
- Department of Molecular Biology, Umeå UniversityUmeåSweden
| | - István Andó
- Biological Research Centre, Institute of Genetics, Innate Immunity Group, Eötvös Loránd Research NetworkSzegedHungary
| |
Collapse
|
21
|
Yu S, Luo F, Xu Y, Zhang Y, Jin LH. Drosophila Innate Immunity Involves Multiple Signaling Pathways and Coordinated Communication Between Different Tissues. Front Immunol 2022; 13:905370. [PMID: 35911716 PMCID: PMC9336466 DOI: 10.3389/fimmu.2022.905370] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune response provides the first line of defense against invading pathogens, and immune disorders cause a variety of diseases. The fruit fly Drosophila melanogaster employs multiple innate immune reactions to resist infection. First, epithelial tissues function as physical barriers to prevent pathogen invasion. In addition, macrophage-like plasmatocytes eliminate intruders through phagocytosis, and lamellocytes encapsulate large particles, such as wasp eggs, that cannot be phagocytosed. Regarding humoral immune responses, the fat body, equivalent to the mammalian liver, secretes antimicrobial peptides into hemolymph, killing bacteria and fungi. Drosophila has been shown to be a powerful in vivo model for studying the mechanism of innate immunity and host-pathogen interactions because Drosophila and higher organisms share conserved signaling pathways and factors. Moreover, the ease with which Drosophila genetic and physiological characteristics can be manipulated prevents interference by adaptive immunity. In this review, we discuss the signaling pathways activated in Drosophila innate immunity, namely, the Toll, Imd, JNK, JAK/STAT pathways, and other factors, as well as relevant regulatory networks. We also review the mechanisms by which different tissues, including hemocytes, the fat body, the lymph gland, muscles, the gut and the brain coordinate innate immune responses. Furthermore, the latest studies in this field are outlined in this review. In summary, understanding the mechanism underlying innate immunity orchestration in Drosophila will help us better study human innate immunity-related diseases.
Collapse
|
22
|
Bakopoulos D, Whisstock JC, Warr CG, Johnson TK. Macrophage self‐renewal is regulated by transient expression of
PDGF‐ and VEGF‐related factor 2. FEBS J 2022; 289:3735-3751. [DOI: 10.1111/febs.16364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/24/2021] [Accepted: 01/19/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Daniel Bakopoulos
- School of Biological Sciences Monash University Clayton Vic. Australia
| | - James C. Whisstock
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University Clayton Vic. Australia
- Department of Biochemistry and Molecular Biology Monash University Clayton Vic. Australia
| | - Coral G. Warr
- School of Biological Sciences Monash University Clayton Vic. Australia
- School of Molecular Sciences La Trobe University Bundoora Vic. Australia
| | - Travis K. Johnson
- School of Biological Sciences Monash University Clayton Vic. Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University Clayton Vic. Australia
| |
Collapse
|
23
|
Hanson MA, Kondo S, Lemaitre B. Drosophila immunity: the Drosocin gene encodes two host defence peptides with pathogen-specific roles. Proc Biol Sci 2022; 289:20220773. [PMID: 35730150 PMCID: PMC9233930 DOI: 10.1098/rspb.2022.0773] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key to defence against infection in plants and animals. Use of AMP mutations in Drosophila has now revealed that AMPs can additively or synergistically contribute to defence in vivo. However, these studies also revealed high specificity, wherein just one AMP contributes an outsized role in combatting a specific pathogen. Here, we show the Drosocin locus (CG10816) is more complex than previously described. In addition to its namesake peptide 'Drosocin', it encodes a second mature peptide from a precursor via furin cleavage. This peptide corresponds to the previously uncharacterized 'Immune-induced Molecule 7'. A polymorphism (Thr52Ala) in the Drosocin precursor protein previously masked the identification of this peptide, which we name 'Buletin'. Using mutations differently affecting Drosocin and Buletin, we show that only Drosocin contributes to Drosocin gene-mediated defence against Enterobacter cloacae. Strikingly, we observed that Buletin, but not Drosocin, contributes to the Drosocin gene-mediated defence against Providencia burhodogranariea, including an importance of the Thr52Ala polymorphism for survival. Our study reveals that the Drosocin gene encodes two prominent host defence peptides with different specificity against distinct pathogens. This finding emphasizes the complexity of the Drosophila humoral response and demonstrates how natural polymorphisms can affect host susceptibility.
Collapse
Affiliation(s)
- M. A. Hanson
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - S. Kondo
- Invertebrate Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - B. Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
24
|
Caravello G, Franchet A, Niehus S, Ferrandon D. Phagocytosis Is the Sole Arm of Drosophila melanogaster Known Host Defenses That Provides Some Protection Against Microsporidia Infection. Front Immunol 2022; 13:858360. [PMID: 35493511 PMCID: PMC9043853 DOI: 10.3389/fimmu.2022.858360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Microsporidia are obligate intracellular parasites able to infest specifically a large range of species, including insects. The knowledge about the biology of microsporidial infections remains confined to mostly descriptive studies, including molecular approaches such as transcriptomics or proteomics. Thus, functional data to understand insect host defenses are currently lacking. Here, we have undertaken a genetic analysis of known host defenses of the Drosophila melanogaster using an infection model whereby Tubulinosema ratisbonensis spores are directly injected in this insect. We find that phagocytosis does confer some protection in this infection model. In contrast, the systemic immune response, extracellular reactive oxygen species, thioester proteins, xenophagy, and intracellular antiviral response pathways do not appear to be involved in the resistance against this parasite. Unexpectedly, several genes such as PGRP-LE seem to promote this infection. The prophenol oxidases that mediate melanization have different functions; PPO1 presents a phenotype similar to that of PGRP-LE whereas that of PPO2 suggests a function in the resilience to infection. Similarly, eiger and Unpaired3, which encode two cytokines secreted by hemocytes display a resilience phenotype with a strong susceptibility to T. ratisbonensis.
Collapse
Affiliation(s)
| | | | | | - Dominique Ferrandon
- UPR9022, University of Strasbourg, Institut de Biologie Moléculaire et Cellulaire (IBMC), Modèles Insectes D’Immunité Innée (M3I) Unité Propre Recherche (UPR) 9022 du Centre National de la Recherche Scientifique (CNRS), Strasbourg, France
| |
Collapse
|
25
|
Koranteng F, Cho B, Shim J. Intrinsic and Extrinsic Regulation of Hematopoiesis in Drosophila. Mol Cells 2022; 45:101-108. [PMID: 35253654 PMCID: PMC8926866 DOI: 10.14348/molcells.2022.2039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022] Open
Abstract
Drosophila melanogaster lymph gland, the primary site of hematopoiesis, contains myeloid-like progenitor cells that differentiate into functional hemocytes in the circulation of pupae and adults. Fly hemocytes are dynamic and plastic, and they play diverse roles in the innate immune response and wound healing. Various hematopoietic regulators in the lymph gland ensure the developmental and functional balance between progenitors and mature blood cells. In addition, systemic factors, such as nutrient availability and sensory inputs, integrate environmental variabilities to synchronize the blood development in the lymph gland with larval growth, physiology, and immunity. This review examines the intrinsic and extrinsic factors determining the progenitor states during hemocyte development in the lymph gland and provides new insights for further studies that may extend the frontier of our collective knowledge on hematopoiesis and innate immunity.
Collapse
Affiliation(s)
| | - Bumsik Cho
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Jiwon Shim
- Department of Life Science, Hanyang University, Seoul 04763, Korea
- Research Institute for Natural Science, Hanyang University, Seoul 04763, Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
26
|
Li H, Janssens J, De Waegeneer M, Kolluru SS, Davie K, Gardeux V, Saelens W, David F, Brbić M, Spanier K, Leskovec J, McLaughlin CN, Xie Q, Jones RC, Brueckner K, Shim J, Tattikota SG, Schnorrer F, Rust K, Nystul TG, Carvalho-Santos Z, Ribeiro C, Pal S, Mahadevaraju S, Przytycka TM, Allen AM, Goodwin SF, Berry CW, Fuller MT, White-Cooper H, Matunis EL, DiNardo S, Galenza A, O’Brien LE, Dow JAT, Jasper H, Oliver B, Perrimon N, Deplancke B, Quake SR, Luo L, Aerts S. Fly Cell Atlas: A single-nucleus transcriptomic atlas of the adult fruit fly. Science 2022; 375:eabk2432. [PMID: 35239393 PMCID: PMC8944923 DOI: 10.1126/science.abk2432] [Citation(s) in RCA: 278] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.
Collapse
Affiliation(s)
- Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jasper Janssens
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium
- Laboratory of Computational Biology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Maxime De Waegeneer
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium
- Laboratory of Computational Biology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Sai Saroja Kolluru
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford CA USA, and Chan Zuckerberg Biohub, San Francisco CA, USA
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium
| | - Vincent Gardeux
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Wouter Saelens
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Fabrice David
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Maria Brbić
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA, and Chan Zuckerberg Biohub, San Francisco CA, USA
| | - Katina Spanier
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium
- Laboratory of Computational Biology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA, and Chan Zuckerberg Biohub, San Francisco CA, USA
| | - Colleen N. McLaughlin
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Robert C. Jones
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford CA USA, and Chan Zuckerberg Biohub, San Francisco CA, USA
| | - Katja Brueckner
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA
| | - Jiwon Shim
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea 04763
| | - Sudhir Gopal Tattikota
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115; Howard Hughes Medical Institute, Boston, MA, USA
| | - Frank Schnorrer
- Aix-Marseille University, CNRS, IBDM (UMR 7288), Turing Centre for Living systems, 13009 Marseille, France
| | - Katja Rust
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University, Marburg, Germany
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Todd G. Nystul
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Zita Carvalho-Santos
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Carlos Ribeiro
- Behavior and Metabolism Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Soumitra Pal
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD 20894, USA
| | - Sharvani Mahadevaraju
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Kidney and Digestive Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Teresa M. Przytycka
- National Center of Biotechnology Information, National Library of Medicine, NIH, Bethesda, MD 20894, USA
| | - Aaron M. Allen
- Centre for Neural Circuits & Behaviour, University of Oxford, Tinsley Building, Mansfield road, Oxford, OX1 3SR, UK
| | - Stephen F. Goodwin
- Centre for Neural Circuits & Behaviour, University of Oxford, Tinsley Building, Mansfield road, Oxford, OX1 3SR, UK
| | - Cameron W. Berry
- Department of Developmental Biology and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret T. Fuller
- Department of Developmental Biology and Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Helen White-Cooper
- Molecular Biosciences Division, Cardiff University, Cardiff, CF10 3AX UK
| | - Erika L. Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen DiNardo
- Perelman School of Medicine, The University of Pennsylvania, and The Penn Institute for Regenerative Medicine Philadelphia, PA 19104, USA
| | - Anthony Galenza
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305, USA
| | - Lucy Erin O’Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford CA 94305, USA
| | - Julian A. T. Dow
- Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - FCA Consortium
- FCA Consortium: All authors listed before Acknowledgements, and all contributions and affiliations listed in the Supplementary Materials
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Brian Oliver
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Kidney and Digestive Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115; Howard Hughes Medical Institute, Boston, MA, USA
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Stephen R. Quake
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford CA USA, and Chan Zuckerberg Biohub, San Francisco CA, USA
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Stein Aerts
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium
- Laboratory of Computational Biology, Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
27
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
28
|
Neuron-derived neuropeptide Y fine-tunes the splenic immune responses. Neuron 2022; 110:1327-1339.e6. [PMID: 35139365 DOI: 10.1016/j.neuron.2022.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/14/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022]
Abstract
The nervous and immune systems are closely entwined to maintain the immune balance in health and disease. Here, we showed that LPS can activate suprarenal and celiac ganglia (SrG-CG) neurons and upregulate NPY expression in rats. Single-cell sequencing analysis revealed that knockdown of the NPY gene in SrG-CG altered the proliferation and activation of splenic lymphocytes. In a neuron and splenocyte coculture system and in vivo experiments, neuronal NPY in SrG-CG attenuated the splenic immune response. Notably, we demonstrated that neuronal NPF in Drosophila exerted a conservative immunomodulatory effect. Moreover, numerous SNPs in NPY and its receptors were significantly associated with human autoimmune diseases, which was further supported by the autoimmune disease patients and mouse model experiments. Together, we demonstrated that NPY is an ancient language for nervous-immune system crosstalk and might be utilized to alleviate inflammatory storms during infection and to modulate immune balance in autoimmune diseases.
Collapse
|
29
|
Sensing microbial infections in the Drosophila melanogaster genetic model organism. Immunogenetics 2022; 74:35-62. [DOI: 10.1007/s00251-021-01239-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022]
|
30
|
Belyaeva V, Wachner S, Gyoergy A, Emtenani S, Gridchyn I, Akhmanova M, Linder M, Roblek M, Sibilia M, Siekhaus D. Fos regulates macrophage infiltration against surrounding tissue resistance by a cortical actin-based mechanism in Drosophila. PLoS Biol 2022; 20:e3001494. [PMID: 34990456 PMCID: PMC8735623 DOI: 10.1371/journal.pbio.3001494] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
The infiltration of immune cells into tissues underlies the establishment of tissue-resident macrophages and responses to infections and tumors. Yet the mechanisms immune cells utilize to negotiate tissue barriers in living organisms are not well understood, and a role for cortical actin has not been examined. Here, we find that the tissue invasion of Drosophila macrophages, also known as plasmatocytes or hemocytes, utilizes enhanced cortical F-actin levels stimulated by the Drosophila member of the fos proto oncogene transcription factor family (Dfos, Kayak). RNA sequencing analysis and live imaging show that Dfos enhances F-actin levels around the entire macrophage surface by increasing mRNA levels of the membrane spanning molecular scaffold tetraspanin TM4SF, and the actin cross-linking filamin Cheerio, which are themselves required for invasion. Both the filamin and the tetraspanin enhance the cortical activity of Rho1 and the formin Diaphanous and thus the assembly of cortical actin, which is a critical function since expressing a dominant active form of Diaphanous can rescue the Dfos macrophage invasion defect. In vivo imaging shows that Dfos enhances the efficiency of the initial phases of macrophage tissue entry. Genetic evidence argues that this Dfos-induced program in macrophages counteracts the constraint produced by the tension of surrounding tissues and buffers the properties of the macrophage nucleus from affecting tissue entry. We thus identify strengthening the cortical actin cytoskeleton through Dfos as a key process allowing efficient forward movement of an immune cell into surrounding tissues. The infiltration of immune cells into tissue underlies the establishment of tissue-resident macrophages, and responses to infections and tumors, but how do they overcome tissue barriers? This study shows that macrophages upregulate the proto-oncogene Fos, increasing the density and crosslinking of cortical actin, thereby counteracting the tension of surrounding tissues and protecting the macrophage nucleus.
Collapse
Affiliation(s)
- Vera Belyaeva
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Stephanie Wachner
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Attila Gyoergy
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Shamsi Emtenani
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Igor Gridchyn
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Maria Akhmanova
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Markus Linder
- Institute of Cancer Research, Department of Medicine 1, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Marko Roblek
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine 1, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daria Siekhaus
- Institute of Science and Technology Austria, Klosterneuburg, Austria
- * E-mail:
| |
Collapse
|
31
|
Dhankhar J, Agrawal N, Shrivastava A. Pan-neuronal expression of human mutant huntingtin protein in Drosophila impairs immune response of hemocytes. J Neuroimmunol 2021; 363:577801. [PMID: 34973473 DOI: 10.1016/j.jneuroim.2021.577801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/19/2022]
Abstract
Huntington's disease (HD) is a late-onset; progressive, dominantly inherited neurological disorder marked by an abnormal expansion of polyglutamine (poly Q) repeats in Huntingtin (HTT) protein. The pathological effects of mutant Huntingtin (mHTT) are not restricted to the nervous system but systemic abnormalities including immune dysregulation have been evidenced in clinical and experimental settings of HD. Indeed, mHTT is ubiquitously expressed and could induce cellular toxicity by directly acting on immune cells. However, it is still unclear if selective expression of mHTT exon1 in neurons could induce immune responses and hemocytes' function. In the present study, we intended to monitor perturbations in the hemocytes' population and their physiological functions in Drosophila, caused by pan-neuronal expression of mHTT protein. A measure of hemocyte count and their physiological activities caused by pan-neuronal expression of mHTT protein highlighted the extent of immune dysregulation occurring with disease progression. We found that pan-neuronal expression of mHTT significantly alters crystal cells and plasmatocyte count in larvae and adults with disease progression. Interestingly, plasmatocytes isolated from diseased conditions exhibit a gradual decline in phagocytic activity ex vivo at progressive stages of the disease as compared to age-matched control groups. In addition, diseased flies displayed elevated reactive oxygen species (ROS) in circulating plasmatocytes at the larval stage and in sessile plasmatocytes of hematopoietic pockets at terminal stages of disease. These findings strongly implicate that neuronal expression of mHTT alone is sufficient to induce non-cell-autonomous immune dysregulation in vivo.
Collapse
Affiliation(s)
- Jyoti Dhankhar
- Department of Zoology, University of Delhi, New Delhi 110007, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi, New Delhi 110007, India.
| | - Anju Shrivastava
- Department of Zoology, University of Delhi, New Delhi 110007, India.
| |
Collapse
|
32
|
Zhao X, Karpac J. Glutamate metabolism directs energetic trade-offs to shape host-pathogen susceptibility in Drosophila. Cell Metab 2021; 33:2428-2444.e8. [PMID: 34710355 PMCID: PMC9153082 DOI: 10.1016/j.cmet.2021.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/26/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022]
Abstract
Individual hosts within populations often show inter-individual variation in their susceptibility to bacterial pathogen-related diseases. Utilizing Drosophila, we highlight that phenotypic variation in host-pathogen susceptibility within populations is driven by energetic trade-offs, facilitated by infection-mediated changes in glutamate metabolism. Furthermore, host-pathogen susceptibility is conditioned by life history, which adjusts immunometabolic sensing in muscles to direct vitamin-dependent reallocation of host energy substrates from the adipose tissue (i.e., a muscle-adipose tissue axis). Life history conditions inter-individual variation in the activation strength of intra-muscular NF-κB signaling. Limited intra-muscular NF-κB signaling activity allows for enhanced infection-mediated mitochondrial biogenesis and function, which stimulates glutamate dehydrogenase-dependent synthesis of glutamate. Muscle-derived glutamate acts as a systemic metabolite to promote lipid mobilization through modulating vitamin B enzymatic cofactor transport and function in the adipose tissue. This energy substrate reallocation improves pathogen clearance and boosts host survival. Finally, life history events that adjust energetic trade-offs can shape inter-individual variation in host-pathogen susceptibility after infection.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Molecular and Cellular Medicine, Texas A&M University, College of Medicine, Bryan, TX 77807, USA
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, Texas A&M University, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
33
|
Rodrigues D, VijayRaghavan K, Waltzer L, Inamdar MS. Intact in situ Preparation of the Drosophila melanogaster Lymph Gland for a Comprehensive Analysis of Larval Hematopoiesis. Bio Protoc 2021; 11:e4204. [PMID: 34859119 DOI: 10.21769/bioprotoc.4204] [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: 04/19/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 11/02/2022] Open
Abstract
Blood cells have a limited lifespan and are replenished by a small number of hematopoietic stem and progenitor cells (HSPCs). Adult vertebrate hematopoiesis occurs in the bone marrow, liver, and spleen, rendering a comprehensive analysis of the entire HSPC pool nearly impossible. The Drosophila blood system is well studied and has developmental, molecular, and functional parallels with that of vertebrates. Unlike vertebrates, post-embryonic hematopoiesis in Drosophila is essentially restricted to the larval lymph gland (LG), a multi-lobed organ that flanks the dorsal vessel. Because the anterior-most or primary lobes of the LG are easy to dissect out, their cellular and molecular characteristics have been studied in considerable detail. The 2-3 pairs of posterior lobes are more delicate and fragile and have largely been ignored. However, posterior lobes harbor a significant blood progenitor pool, and several hematopoietic mutants show differences in phenotype between the anterior and posterior lobes. Hence, a comprehensive analysis of the LG is important for a thorough understanding of Drosophila hematopoiesis. Most studies focus on isolating the primary lobes by methods that generally dislodge and damage other lobes. To obtain preparations of the whole LG, including intact posterior lobes, here we provide a detailed protocol for larval fillet dissection. This allows accessing and analyzing complete LG lobes, along with dorsal vessel and pericardial cells. We demonstrate that tissue architecture and integrity is maintained and provide methods for quantitative analysis. This protocol can be used to quickly and effectively isolate complete LGs from first instar larval to pupal stages and can be implemented with ease.
Collapse
Affiliation(s)
- Diana Rodrigues
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.,National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology and Research Academy, Tamil Nadu, India
| | - K VijayRaghavan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology and Research Academy, Tamil Nadu, India
| | - Lucas Waltzer
- University of Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - Maneesha S Inamdar
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
34
|
Boulet M, Renaud Y, Lapraz F, Benmimoun B, Vandel L, Waltzer L. Characterization of the Drosophila Adult Hematopoietic System Reveals a Rare Cell Population With Differentiation and Proliferation Potential. Front Cell Dev Biol 2021; 9:739357. [PMID: 34722521 PMCID: PMC8550105 DOI: 10.3389/fcell.2021.739357] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023] Open
Abstract
While many studies have described Drosophila embryonic and larval blood cells, the hematopoietic system of the imago remains poorly characterized and conflicting data have been published concerning adult hematopoiesis. Using a combination of blood cell markers, we show that the adult hematopoietic system is essentially composed of a few distinct mature blood cell types. In addition, our transcriptomics results indicate that adult and larval blood cells have both common and specific features and it appears that adult hemocytes reactivate many genes expressed in embryonic blood cells. Interestingly, we identify a small set of blood cells that does not express differentiation markers but rather maintains the expression of the progenitor marker domeMeso. Yet, we show that these cells are derived from the posterior signaling center, a specialized population of cells present in the larval lymph gland, rather than from larval blood cell progenitors, and that their maintenance depends on the EBF transcription factor Collier. Furthermore, while these cells are normally quiescent, we find that some of them can differentiate and proliferate in response to bacterial infection. In sum, our results indicate that adult flies harbor a small population of specialized cells with limited hematopoietic potential and further support the idea that no substantial hematopoiesis takes place during adulthood.
Collapse
Affiliation(s)
- Manon Boulet
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - Yoan Renaud
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - François Lapraz
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Billel Benmimoun
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| | - Laurence Vandel
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France
| | - Lucas Waltzer
- Université Clermont Auvergne, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Medicale, Institut Génétique Reproduction et Développement, Clermont-Ferrand, France.,Centre de Biologie du Développement, Centre de Biologie Intégrative, Université de Toulouse, Centre National de la Recherche Scientifique, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
35
|
Bilder D, Ong K, Hsi TC, Adiga K, Kim J. Tumour-host interactions through the lens of Drosophila. Nat Rev Cancer 2021; 21:687-700. [PMID: 34389815 PMCID: PMC8669834 DOI: 10.1038/s41568-021-00387-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
There is a large gap between the deep understanding of mechanisms driving tumour growth and the reasons why patients ultimately die of cancer. It is now appreciated that interactions between the tumour and surrounding non-tumour (sometimes referred to as host) cells play critical roles in mortality as well as tumour progression, but much remains unknown about the underlying molecular mechanisms, especially those that act beyond the tumour microenvironment. Drosophila has a track record of high-impact discoveries about cell-autonomous growth regulation, and is well suited to now probe mysteries of tumour - host interactions. Here, we review current knowledge about how fly tumours interact with microenvironmental stroma, circulating innate immune cells and distant organs to influence disease progression. We also discuss reciprocal regulation between tumours and host physiology, with a particular focus on paraneoplasias. The fly's simplicity along with the ability to study lethality directly provide an opportunity to shed new light on how cancer actually kills.
Collapse
Affiliation(s)
- David Bilder
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| | - Katy Ong
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Tsai-Ching Hsi
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Kavya Adiga
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jung Kim
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| |
Collapse
|
36
|
Morin-Poulard I, Tian Y, Vanzo N, Crozatier M. Drosophila as a Model to Study Cellular Communication Between the Hematopoietic Niche and Blood Progenitors Under Homeostatic Conditions and in Response to an Immune Stress. Front Immunol 2021; 12:719349. [PMID: 34484226 PMCID: PMC8415499 DOI: 10.3389/fimmu.2021.719349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 12/23/2022] Open
Abstract
In adult mammals, blood cells are formed from hematopoietic stem progenitor cells, which are controlled by a complex cellular microenvironment called "niche". Drosophila melanogaster is a powerful model organism to decipher the mechanisms controlling hematopoiesis, due both to its limited number of blood cell lineages and to the conservation of genes and signaling pathways throughout bilaterian evolution. Insect blood cells or hemocytes are similar to the mammalian myeloid lineage that ensures innate immunity functions. Like in vertebrates, two waves of hematopoiesis occur in Drosophila. The first wave takes place during embryogenesis. The second wave occurs at larval stages, where two distinct hematopoietic sites are identified: subcuticular hematopoietic pockets and a specialized hematopoietic organ called the lymph gland. In both sites, hematopoiesis is regulated by distinct niches. In hematopoietic pockets, sensory neurons of the peripheral nervous system provide a microenvironment that promotes embryonic hemocyte expansion and differentiation. In the lymph gland blood cells are produced from hematopoietic progenitors. A small cluster of cells called Posterior Signaling Centre (PSC) and the vascular system, along which the lymph gland develops, act collectively as a niche, under homeostatic conditions, to control the balance between maintenance and differentiation of lymph gland progenitors. In response to an immune stress such as wasp parasitism, lymph gland hematopoiesis is drastically modified and shifts towards emergency hematopoiesis, leading to increased progenitor proliferation and their differentiation into lamellocyte, a specific blood cell type which will neutralize the parasite. The PSC is essential to control this emergency response. In this review, we summarize Drosophila cellular and molecular mechanisms involved in the communication between the niche and hematopoietic progenitors, both under homeostatic and stress conditions. Finally, we discuss similarities between mechanisms by which niches regulate hematopoietic stem/progenitor cells in Drosophila and mammals.
Collapse
Affiliation(s)
| | - Yushun Tian
- MCD/UMR5077, Centre de Biologie Intégrative (CBI), Toulouse, France
| | - Nathalie Vanzo
- MCD/UMR5077, Centre de Biologie Intégrative (CBI), Toulouse, France
| | | |
Collapse
|
37
|
Kaech H, Dennis AB, Vorburger C. Triple RNA-Seq characterizes aphid gene expression in response to infection with unequally virulent strains of the endosymbiont Hamiltonella defensa. BMC Genomics 2021; 22:449. [PMID: 34134631 PMCID: PMC8207614 DOI: 10.1186/s12864-021-07742-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/20/2021] [Indexed: 11/10/2022] Open
Abstract
Background Secondary endosymbionts of aphids provide benefits to their hosts, but also impose costs such as reduced lifespan and reproductive output. The aphid Aphis fabae is host to different strains of the secondary endosymbiont Hamiltonella defensa, which encode different putative toxins. These strains have very different phenotypes: They reach different densities in the host, and the costs and benefits (protection against parasitoid wasps) they confer to the host vary strongly. Results We used RNA-Seq to generate hypotheses on why four of these strains inflict such different costs to A. fabae. We found different H. defensa strains to cause strain-specific changes in aphid gene expression, but little effect of H. defensa on gene expression of the primary endosymbiont, Buchnera aphidicola. The highly costly and over-replicating H. defensa strain H85 was associated with strongly reduced aphid expression of hemocytin, a marker of hemocytes in Drosophila. The closely related strain H15 was associated with downregulation of ubiquitin-related modifier 1, which is related to nutrient-sensing and oxidative stress in other organisms. Strain H402 was associated with strong differential regulation of a set of hypothetical proteins, the majority of which were only differentially regulated in presence of H402. Conclusions Overall, our results suggest that costs of different strains of H. defensa are likely caused by different mechanisms, and that these costs are imposed by interacting with the host rather than the host’s obligatory endosymbiont B. aphidicola. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07742-8.
Collapse
Affiliation(s)
- Heidi Kaech
- Aquatic Ecology, Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland. .,D-USYS, Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland.
| | - Alice B Dennis
- Institute of Biochemistry and Biology, University Potsdam, Potsdam, Germany
| | - Christoph Vorburger
- Aquatic Ecology, Eawag, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, Switzerland.,D-USYS, Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
38
|
Leite THJF, Ferreira ÁGA, Imler JL, Marques JT. Distinct Roles of Hemocytes at Different Stages of Infection by Dengue and Zika Viruses in Aedes aegypti Mosquitoes. Front Immunol 2021; 12:660873. [PMID: 34093550 PMCID: PMC8169962 DOI: 10.3389/fimmu.2021.660873] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
Aedes aegypti mosquitoes are vectors for arboviruses of medical importance such as dengue (DENV) and Zika (ZIKV) viruses. Different innate immune pathways contribute to the control of arboviruses in the mosquito vector including RNA interference, Toll and Jak-STAT pathways. However, the role of cellular responses mediated by circulating macrophage-like cells known as hemocytes remains unclear. Here we show that hemocytes are recruited to the midgut of Ae. aegypti mosquitoes in response to DENV or ZIKV. Blockade of the phagocytic function of hemocytes using latex beads induced increased accumulation of hemocytes in the midgut and a reduction in virus infection levels in this organ. In contrast, inhibition of phagocytosis by hemocytes led to increased systemic dissemination and replication of DENV and ZIKV. Hence, our work reveals a dual role for hemocytes in Ae. aegypti mosquitoes, whereby phagocytosis is not required to control viral infection in the midgut but is essential to restrict systemic dissemination. Further understanding of the mechanism behind this duality could help the design of vector-based strategies to prevent transmission of arboviruses.
Collapse
Affiliation(s)
- Thiago H J F Leite
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Álvaro G A Ferreira
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Brazil
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| | - João T Marques
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Université de Strasbourg, CNRS UPR9022, Inserm U1257, Strasbourg, France
| |
Collapse
|
39
|
Coates JA, Brooks E, Brittle AL, Armitage EL, Zeidler MP, Evans IR. Identification of functionally distinct macrophage subpopulations in Drosophila. eLife 2021; 10:e58686. [PMID: 33885361 PMCID: PMC8062135 DOI: 10.7554/elife.58686] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Vertebrate macrophages are a highly heterogeneous cell population, but while Drosophila blood is dominated by a macrophage-like lineage (plasmatocytes), until very recently these cells were considered to represent a homogeneous population. Here, we present our identification of enhancer elements labelling plasmatocyte subpopulations, which vary in abundance across development. These subpopulations exhibit functional differences compared to the overall population, including more potent injury responses and differential localisation and dynamics in pupae and adults. Our enhancer analysis identified candidate genes regulating plasmatocyte behaviour: pan-plasmatocyte expression of one such gene (Calnexin14D) improves wound responses, causing the overall population to resemble more closely the subpopulation marked by the Calnexin14D-associated enhancer. Finally, we show that exposure to increased levels of apoptotic cell death modulates subpopulation cell numbers. Taken together this demonstrates macrophage heterogeneity in Drosophila, identifies mechanisms involved in subpopulation specification and function and facilitates the use of Drosophila to study macrophage heterogeneity in vivo.
Collapse
Affiliation(s)
- Jonathon Alexis Coates
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Elliot Brooks
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Amy Louise Brittle
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Emma Louise Armitage
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Martin Peter Zeidler
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Iwan Robert Evans
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
40
|
Mase A, Augsburger J, Brückner K. Macrophages and Their Organ Locations Shape Each Other in Development and Homeostasis - A Drosophila Perspective. Front Cell Dev Biol 2021; 9:630272. [PMID: 33777939 PMCID: PMC7991785 DOI: 10.3389/fcell.2021.630272] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Across the animal kingdom, macrophages are known for their functions in innate immunity, but they also play key roles in development and homeostasis. Recent insights from single cell profiling and other approaches in the invertebrate model organism Drosophila melanogaster reveal substantial diversity among Drosophila macrophages (plasmatocytes). Together with vertebrate studies that show genuine expression signatures of macrophages based on their organ microenvironments, it is expected that Drosophila macrophage functional diversity is shaped by their anatomical locations and systemic conditions. In vivo evidence for diverse macrophage functions has already been well established by Drosophila genetics: Drosophila macrophages play key roles in various aspects of development and organogenesis, including embryogenesis and development of the nervous, digestive, and reproductive systems. Macrophages further maintain homeostasis in various organ systems and promote regeneration following organ damage and injury. The interdependence and interplay of tissues and their local macrophage populations in Drosophila have implications for understanding principles of organ development and homeostasis in a wide range of species.
Collapse
Affiliation(s)
- Anjeli Mase
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jordan Augsburger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Katja Brückner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
41
|
Arroyo Portilla C, Tomas J, Gorvel JP, Lelouard H. From Species to Regional and Local Specialization of Intestinal Macrophages. Front Cell Dev Biol 2021; 8:624213. [PMID: 33681185 PMCID: PMC7930007 DOI: 10.3389/fcell.2020.624213] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Initially intended for nutrient uptake, phagocytosis represents a central mechanism of debris removal and host defense against invading pathogens through the entire animal kingdom. In vertebrates and also many invertebrates, macrophages (MFs) and MF-like cells (e.g., coelomocytes and hemocytes) are professional phagocytic cells that seed tissues to maintain homeostasis through pathogen killing, efferocytosis and tissue shaping, repair, and remodeling. Some MF functions are common to all species and tissues, whereas others are specific to their homing tissue. Indeed, shaped by their microenvironment, MFs become adapted to perform particular functions, highlighting their great plasticity and giving rise to high population diversity. Interestingly, the gut displays several anatomic and functional compartments with large pools of strikingly diversified MF populations. This review focuses on recent advances on intestinal MFs in several species, which have allowed to infer their specificity and functions.
Collapse
Affiliation(s)
- Cynthia Arroyo Portilla
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Departamento de Análisis Clínicos, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Julie Tomas
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | |
Collapse
|
42
|
Rodrigues D, Renaud Y, VijayRaghavan K, Waltzer L, Inamdar MS. Differential activation of JAK-STAT signaling reveals functional compartmentalization in Drosophila blood progenitors. eLife 2021; 10:61409. [PMID: 33594977 PMCID: PMC7920551 DOI: 10.7554/elife.61409] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Blood cells arise from diverse pools of stem and progenitor cells. Understanding progenitor heterogeneity is a major challenge. The Drosophila larval lymph gland is a well-studied model to understand blood progenitor maintenance and recapitulates several aspects of vertebrate hematopoiesis. However in-depth analysis has focused on the anterior lobe progenitors (AP), ignoring the posterior progenitors (PP) from the posterior lobes. Using in situ expression mapping and developmental and transcriptome analysis, we reveal PP heterogeneity and identify molecular-genetic tools to study this abundant progenitor population. Functional analysis shows that PP resist differentiation upon immune challenge, in a JAK-STAT-dependent manner. Upon wasp parasitism, AP downregulate JAK-STAT signaling and form lamellocytes. In contrast, we show that PP activate STAT92E and remain undifferentiated, promoting survival. Stat92E knockdown or genetically reducing JAK-STAT signaling permits PP lamellocyte differentiation. We discuss how heterogeneity and compartmentalization allow functional segregation in response to systemic cues and could be widely applicable.
Collapse
Affiliation(s)
- Diana Rodrigues
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.,National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology & Research Academy, Tamil Nadu, India
| | - Yoan Renaud
- University of Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - K VijayRaghavan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Shanmugha Arts, Science, Technology & Research Academy, Tamil Nadu, India
| | - Lucas Waltzer
- University of Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - Maneesha S Inamdar
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| |
Collapse
|
43
|
Yu S, Luo F, Jin LH. Rab5 and Rab11 maintain hematopoietic homeostasis by restricting multiple signaling pathways in Drosophila. eLife 2021; 10:60870. [PMID: 33560224 PMCID: PMC7891935 DOI: 10.7554/elife.60870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 02/08/2021] [Indexed: 12/26/2022] Open
Abstract
The hematopoietic system of Drosophila is a powerful genetic model for studying hematopoiesis, and vesicle trafficking is important for signal transduction during various developmental processes; however, its interaction with hematopoiesis is currently largely unknown. In this article, we selected three endosome markers, Rab5, Rab7, and Rab11, that play a key role in membrane trafficking and determined whether they participate in hematopoiesis. Inhibiting Rab5 or Rab11 in hemocytes or the cortical zone (CZ) significantly induced cell overproliferation and lamellocyte formation in circulating hemocytes and lymph glands and disrupted blood cell progenitor maintenance. Lamellocyte formation involves the JNK, Toll, and Ras/EGFR signaling pathways. Notably, lamellocyte formation was also associated with JNK-dependent autophagy. In conclusion, we identified Rab5 and Rab11 as novel regulators of hematopoiesis, and our results advance the understanding of the mechanisms underlying the maintenance of hematopoietic homeostasis as well as the pathology of blood disorders such as leukemia.
Collapse
Affiliation(s)
- Shichao Yu
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Fangzhou Luo
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry University, Harbin, China
| |
Collapse
|
44
|
Rodrigues D, VijayRaghavan K, Waltzer L, Inamdar M. Intact in situ Preparation of Drosophila melanogaster Lymph Gland for Comprehensive Analysis of Larval Hematopoiesis. Bio Protoc 2021. [DOI: 10.21769/bioprotoc.4052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
45
|
Lan W, Liu S, Zhao L, Su Y. Regulation of Drosophila Hematopoiesis in Lymph Gland: From a Developmental Signaling Point of View. Int J Mol Sci 2020; 21:ijms21155246. [PMID: 32722007 PMCID: PMC7432643 DOI: 10.3390/ijms21155246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The Drosophila hematopoietic system is becoming increasingly attractive for its simple blood cell lineage and its developmental and functional parallels with the vertebrate system. As the dedicated organ for Drosophila larval hematopoiesis, the lymph gland harbors both multipotent stem-like progenitor cells and differentiated blood cells. The balance between progenitor maintenance and differentiation in the lymph gland must be precisely and tightly controlled. Multiple developmental signaling pathways, such as Notch, Hedgehog, and Wnt/Wingless, have been demonstrated to regulate the hematopoietic processes in the lymph gland. Focusing on blood cell maintenance and differentiation, this article summarizes the functions of several classic developmental signaling pathways for lymph gland growth and patterning, highlighting the important roles of developmental signaling during lymph gland development as well as Drosophila larval hematopoiesis.
Collapse
Affiliation(s)
- Wenwen Lan
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Sumin Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- Fisheries College, Ocean University of China, Qingdao 266003, China
- Correspondence: (L.Z.); (Y.S.)
| | - Ying Su
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (W.L.); (S.L.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Correspondence: (L.Z.); (Y.S.)
| |
Collapse
|
46
|
Insulin-Like Signalling Influences the Coordination of Larval Hemocyte Number with Body Size in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2020; 10:2213-2220. [PMID: 32341056 PMCID: PMC7341137 DOI: 10.1534/g3.120.401313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blood cells, known as hemocytes in invertebrates, play important and conserved roles in immunity, wound healing and tissue remodelling. The control of hemocyte number is therefore critical to ensure these functions are not compromised, and studies using Drosophila melanogaster are proving useful for understanding how this occurs. Recently, the embryonic patterning gene, torso-like (tsl), was identified as being required both for normal hemocyte development and for providing immunity against certain pathogens. Here, we report that Tsl is required specifically during the larval phase of hematopoiesis, and that tsl mutant larvae likely have reduced hemocyte numbers due to a reduced larval growth rate and compromised insulin signaling. Consistent with this, we find that impairing insulin-mediated growth, either by nutrient deprivation or genetically, results in fewer hemocytes. This is likely the result of impaired insulin-like signaling in the hemocytes themselves, since modulation of Insulin-like Receptor (InR) activity specifically in hemocytes causes concomitant changes to their population size in developing larvae. Taken together, our work reveals the strong relationship that exists between body size and hemocyte number, and suggests that insulin-like signaling contributes to, but is not solely responsible for, keeping these tightly aligned during larval development.
Collapse
|
47
|
Abstract
Single-cell RNA sequencing has revealed distinct subpopulations of hemocytes in fruit fly larvae.
Collapse
Affiliation(s)
- Samuel Liegeois
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, UPR 9022 du CNRSStrasbourgFrance
- Sino-French Hoffmann Institute, Guangzhou Medical UniversityGuangzhouChina
| | - Dominique Ferrandon
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, UPR 9022 du CNRSStrasbourgFrance
- Sino-French Hoffmann Institute, Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
48
|
Ramond E, Dudzic JP, Lemaitre B. Comparative RNA-Seq analyses of Drosophila plasmatocytes reveal gene specific signatures in response to clean injury and septic injury. PLoS One 2020; 15:e0235294. [PMID: 32598400 PMCID: PMC7323993 DOI: 10.1371/journal.pone.0235294] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/11/2020] [Indexed: 12/27/2022] Open
Abstract
Drosophila melanogaster's blood cells (hemocytes) play essential roles in wound healing and are involved in clearing microbial infections. Here, we report the transcriptional changes of larval plasmatocytes after clean injury or infection with the Gram-negative bacterium Escherichia coli or the Gram-positive bacterium Staphylococcus aureus compared to hemocytes recovered from unchallenged larvae via RNA-Sequencing. This study reveals 676 differentially expressed genes (DEGs) in hemocytes from clean injury samples compared to unchallenged samples, and 235 and 184 DEGs in E. coli and S. aureus samples respectively compared to clean injury samples. The clean injury samples showed enriched DEGs for immunity, clotting, cytoskeleton, cell migration, hemocyte differentiation, and indicated a metabolic reprogramming to aerobic glycolysis, a well-defined metabolic adaptation observed in mammalian macrophages. Microbial infections trigger significant transcription of immune genes, with significant differences between the E. coli and S. aureus samples suggesting that hemocytes have the ability to engage various programs upon infection. Collectively, our data bring new insights on Drosophila hemocyte function and open the route to post-genomic functional analysis of the cellular immune response.
Collapse
Affiliation(s)
- Elodie Ramond
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jan Paul Dudzic
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
49
|
New insights on Drosophila antimicrobial peptide function in host defense and beyond. Curr Opin Immunol 2020; 62:22-30. [DOI: 10.1016/j.coi.2019.11.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
|
50
|
Belmonte RL, Corbally MK, Duneau DF, Regan JC. Sexual Dimorphisms in Innate Immunity and Responses to Infection in Drosophila melanogaster. Front Immunol 2020; 10:3075. [PMID: 32076419 PMCID: PMC7006818 DOI: 10.3389/fimmu.2019.03075] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022] Open
Abstract
The sexes show profound differences in responses to infection and the development of autoimmunity. Dimorphisms in immune responses are ubiquitous across taxa, from arthropods to vertebrates. Drosophila melanogaster shows strong sex dimorphisms in immune system responses at baseline, upon pathogenic challenge, and over aging. We have performed an exhaustive survey of peer-reviewed literature on Drosophila immunity, and present a database of publications indicating the sex(es) analyzed in each study. While we found a growing interest in the community in adult immunity and in reporting both sexes, the main body of work in this field uses only one sex, or does not stratify by sex. We synthesize evidence for sexually dimorphic responses to bacterial, viral, and fungal infections. Dimorphisms may be mediated by distinct immune compartments, and we review work on sex differences in behavioral, epithelial, cellular, and systemic (fat body-mediated) immunity. Emerging work on sexually dimorphic aging of immune tissues, immune senescence, and inflammation are examined. We consider evolutionary drivers for sex differences in immune investment, highlight the features of Drosophila biology that make it particularly amenable to studies of immune dimorphisms, and discuss areas for future exploration.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary-Kate Corbally
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David F. Duneau
- Laboratoire Evolution & Diversite Biologique, UMR5174 EDB, CNRS, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Jennifer C. Regan
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|