1
|
Glazer I, Simões N, Eleftherianos I, Ramakrishnan J, Ment D, Toubarro D, Mallick S. Entomopathogenic nematodes: Survival, virulence and immunity. J Invertebr Pathol 2025; 212:108363. [PMID: 40412605 DOI: 10.1016/j.jip.2025.108363] [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: 12/31/2024] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
As entomopathogenic nematodes (EPNs) are used as biological control agents, their survival and persistence are crucial to ensure success in application against insect pests. The survival of Heterorhabditis and Steinernema species is dependent on abiotic and biotic factors in the environment. Abiotic stress environments such as desiccation, temperature, and ultraviolet radiation (UV) severely impact their performance on field. EPNs produce and secrete effector molecules during the early stages of infection to interfere with the molecular mechanisms that control the insect innate immune function. Also, EPN effectors facilitate the subsequent release and spread of their symbiotic bacteria within the host. Hence, a comprehensive understanding of the underlying survival and virulence mechanisms enabling protection against environmental conditions and insect host immune responses is imperative to realistically enhance their performance on field. Thus, identifying key players regulating EPN survival, virulence and immunity could invariably contribute towards developing more robust, reliable solutions and application strategies including genetic tools and formulation technologies.
Collapse
Affiliation(s)
- Itamar Glazer
- Institute of Plant Protection, ARO, The Volcani Center, Rishon LeZion, Israel
| | - Nelson Simões
- Biotechnology Centre of Azores (CBA), Faculty of Sciences and Technology, University of the Azores, Açores 9500-321 Ponta Delgada, Portugal
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Lab, Department of Biological Sciences, The George Washington University, Washington, DC, United States.
| | - Jayashree Ramakrishnan
- Institute of Plant Protection, ARO, The Volcani Center, Rishon LeZion, Israel; The Robert H. Smith Faculty of Agriculture, Food & Environment the Hebrew University of Jerusalem, Rehovot, Israel
| | - Dana Ment
- Institute of Plant Protection, ARO, The Volcani Center, Rishon LeZion, Israel
| | - Duarte Toubarro
- Biotechnology Centre of Azores (CBA), Faculty of Sciences and Technology, University of the Azores, Açores 9500-321 Ponta Delgada, Portugal
| | - Sreeradha Mallick
- Infection and Innate Immunity Lab, Department of Biological Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
2
|
Marcetteau J, Duarte P, Leitão AB, Sucena É. Transdifferentiation of plasmatocytes to crystal cells in the lymph gland of Drosophila melanogaster. EMBO Rep 2025; 26:2077-2097. [PMID: 40075235 PMCID: PMC12019564 DOI: 10.1038/s44319-025-00366-z] [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: 02/06/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 03/14/2025] Open
Abstract
Under homeostatic conditions, haematopoiesis in Drosophila larvae occurs in the lymph gland and sessile haemocyte clusters to produce two functionally and morphologically different cells: plasmatocytes and crystal cells. It is well-established that in the lymph gland both cell types stem from a binary decision of the medullary prohaemocyte precursors. However, in sessile clusters and dorsal vessel, crystal cells have been shown to originate from the transdifferentiation of plasmatocytes in a Notch/Serrate-dependent manner. We show that transdifferentiation occurs also in the lymph gland. In vivo phagocytosis assays confirm that cortical plasmatocytes are functionally differentiated phagocytic cells. We uncover a double-positive population in the cortical zone that lineage-tracing and long-term live imaging experiments show will differentiate into crystal cells. The reduction of Notch levels within the lymph gland plasmatocyte population reduces crystal cell number. This extension of a transdifferentiation mechanism reinforces the growing role of haematopoietic plasticity in maintaining homeostasis in Drosophila and vertebrate systems. Future work should test the regulation and relative contribution of these two processes under different immunological and/or metabolic conditions.
Collapse
Affiliation(s)
- Julien Marcetteau
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | - Patrícia Duarte
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal
| | | | - Élio Sucena
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal.
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Edifício C2, Campo Grande, 1749-016, Lisbon, Portugal.
- cE3c: Centre for Ecology, Evolution and Environmental Changes, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisbon, Portugal.
| |
Collapse
|
3
|
Luo W, Zhang F, Zhao F, Fang Y, Zhao L, Su Y. Dual role of PpV in Drosophila crystal cell proliferation and survival. J Mol Cell Biol 2025; 16:mjae028. [PMID: 39085037 PMCID: PMC11927399 DOI: 10.1093/jmcb/mjae028] [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: 01/18/2024] [Revised: 05/28/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024] Open
Abstract
Drosophila melanogaster crystal cells are a specialized type of blood cells for the innate immune process upon injury. Under normal conditions, crystal cells rarely proliferate and constitute a small proportion of fly blood cells. Notch signaling has been known to guide the cell fate determination of crystal cells and maintain their survival. Here, we reported that protein phosphatase V (PpV), the unique catalytic subunit of protein phosphatase 6 in Drosophila, is a novel regulator of crystal cell proliferation and integrity. We found that PpV proteins highly accumulated in crystal cells in the larval hematopoietic organ termed the lymph gland. Silencing PpV using RNA interference led to increased crystal cell proliferation in a Notch-independent manner and induced crystal cell rupture dependent on Notch signaling. Moreover, additive PpV prevented the rupture of crystal cells in lymph glands upon a needle injury, suggesting the involvement of PpV in wound healing. Altogether, our results indicated that PpV plays a dual role in lymph glands, preventing crystal cell proliferation to limit the cell number, as well as inhibiting crystal cell rupture to maintain their survival.
Collapse
Affiliation(s)
- Wang Luo
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Fangzhen Zhao
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yang Fang
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Long Zhao
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Ying Su
- Key Laboratory of Evolution & Marine Biodiversity (Ministry of Education) and Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
4
|
Luo F, Sui L, Sun Y, Lai Z, Zhang C, Zhang G, Bi B, Yu S, Jin LH. Rab1 and Syntaxin 17 regulate hematopoietic homeostasis through β-integrin trafficking in Drosophila. J Genet Genomics 2025; 52:51-65. [PMID: 39542172 DOI: 10.1016/j.jgg.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Hematopoiesis is crucial for organismal health, and Drosophila serves as an effective genetic model due to conserved regulatory mechanisms with vertebrates. In larvae, hematopoiesis primarily occurs in the lymph gland, which contains distinct zones, including the cortical zone, intermediate zone, medullary zone, and posterior signaling center (PSC). Rab1 is vital for membrane trafficking and maintaining the localization of cell adhesion molecules, yet its role in hematopoietic homeostasis is not fully understood. This study investigates the effects of Rab1 dysfunction on β-integrin trafficking within circulating hemocytes and lymph gland cells. Rab1 impairment disrupts the endosomal trafficking of β-integrin, leading to its abnormal localization on cell membranes, which promotes lamellocyte differentiation and alters progenitor dynamics in circulating hemocytes and lymph glands, respectively. We also show that the mislocalization of β-integrin is dependent on the adhesion protein DE-cadherin. The reduction of β-integrin at cell boundaries in PSC cells leads to fewer PSC cells and lamellocyte differentiation. Furthermore, Rab1 regulates the trafficking of β-integrin via the Q-SNARE protein Syntaxin 17 (Syx17). Our findings indicate that Rab1 and Syx17 regulate distinct trafficking pathways for β-integrin in different hematopoietic compartments and maintain hematopoietic homeostasis of Drosophila.
Collapse
Affiliation(s)
- Fangzhou Luo
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Luwei Sui
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Ying Sun
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Zhixian Lai
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Chengcheng Zhang
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Gaoqun Zhang
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Bing Bi
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China
| | - Shichao Yu
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China.
| | - Li Hua Jin
- College of Life Sciences, Northeast Forestry University, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
5
|
Sun LN, Meng JY, Wang Z, Lin SY, Shen J, Yan S. Research progress of aphid immunity system: Potential effective target for green pest management. INSECT SCIENCE 2024; 31:1662-1674. [PMID: 38415382 DOI: 10.1111/1744-7917.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/29/2024]
Abstract
Due to the absence of acquired immunity, insects primarily rely on their innate immune system to resist pathogenic microorganisms and parasitoids in natural habitats. This innate immune system can be classified into cellular immunity and humoral immunity. Cellular immunity is mediated by hemocytes, which perform phagocytosis, aggregation, and encapsulation to fight against invaders, whereas the humoral immunity primarily activates the immune signaling pathways and induces the generation of immune effectors. Existing studies have revealed that the hemipteran aphids lack some crucial immune genes compared to other insect species, indicating the different immune mechanisms in aphids. The current review summarizes the adverse impacts of pathogenic microorganisms and parasitoids on aphids, introduces the cellular and humoral immune systems in insects, and analyzes the differences between aphids and other insect species. Furthermore, our review also discussed the existing prospects and challenges in aphid immunity research, and proposed the potential application of immune genes in green pest management.
Collapse
Affiliation(s)
- Li-Na Sun
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jian-Yu Meng
- Guizhou Tobacco Science Research Institute, Guiyang, China
| | - Zeng Wang
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shi-Yang Lin
- Pu'er Agricultural Science Research Institute, Pu'er, Yunnan Province, China
| | - Jie Shen
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shuo Yan
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
6
|
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
|
7
|
Vincow ES, Thomas RE, Milstein G, Pareek G, Bammler TK, MacDonald J, Pallanck LJ. Glucocerebrosidase deficiency leads to neuropathology via cellular immune activation. PLoS Genet 2024; 20:e1011105. [PMID: 39527642 PMCID: PMC11581407 DOI: 10.1371/journal.pgen.1011105] [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: 12/12/2023] [Revised: 11/21/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Mutations in GBA (glucosylceramidase beta), which encodes the lysosomal enzyme glucocerebrosidase (GCase), are the strongest genetic risk factor for the neurodegenerative disorders Parkinson's disease (PD) and Lewy body dementia. Recent work has suggested that neuroinflammation may be an important factor in the risk conferred by GBA mutations. We therefore systematically tested the contributions of immune-related genes to neuropathology in a Drosophila model of GCase deficiency. We identified target immune factors via RNA-Seq and proteomics on heads from GCase-deficient flies, which revealed both increased abundance of humoral factors and increased macrophage activation. We then manipulated the identified immune factors and measured their effect on head protein aggregates, a hallmark of neurodegenerative disease. Genetic ablation of humoral (secreted) immune factors did not suppress the development of protein aggregation. By contrast, re-expressing Gba1b in activated macrophages suppressed head protein aggregation in Gba1b mutants and rescued their lifespan and behavioral deficits. Moreover, reducing the GCase substrate glucosylceramide in activated macrophages also ameliorated Gba1b mutant phenotypes. Taken together, our findings show that glucosylceramide accumulation due to GCase deficiency leads to macrophage activation, which in turn promotes the development of neuropathology.
Collapse
Affiliation(s)
- Evelyn S. Vincow
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Ruth E. Thomas
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Gillian Milstein
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Gautam Pareek
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Leo J. Pallanck
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
8
|
Samantsidis GR, Smith RC. Exploring new dimensions of immune cell biology in Anopheles gambiae through genetic immunophenotyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619690. [PMID: 39484609 PMCID: PMC11526922 DOI: 10.1101/2024.10.22.619690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mosquito immune cells, or hemocytes, are integral components of the innate immune responses that define vector competence. However, the lack of genetic resources has limited their characterization and our understanding of their functional roles in immune signaling. To overcome these challenges, we engineered transgenic Anopheles gambiae that express fluorescent proteins under the control of candidate hemocyte promoters. Following the characterization of five transgenic constructs through gene expression and microscopy-based approaches, we examine mosquito immune cell populations by leveraging advanced spectral imaging flow cytometry. Our results comprehensively map the composition of mosquito hemocytes, classifying them into twelve distinct populations based on size, granularity, ploidy, phagocytic capacity, and the expression of PPO6, SPARC, and LRIM15 genetic markers. Together, our novel use of morphological properties and genetic markers provides increased resolution into our understanding of mosquito hemocytes, highlighting the complexity and plasticity of these immune cell populations, while providing the foundation for deeper investigations into their roles in immunity and pathogen transmission.
Collapse
Affiliation(s)
| | - Ryan C Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
9
|
Kazek M, Kaczmarek A, Wrońska AK, Boguś MI. Effect of Benzyl Alcohol on Main Defense System Components of Galleria mellonella (Lepidoptera). Int J Mol Sci 2024; 25:11209. [PMID: 39456990 PMCID: PMC11508370 DOI: 10.3390/ijms252011209] [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: 09/11/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Benzyl alcohol (E1519) is an aromatic alcohol used in the pharmaceutical and food industry. It is used to protect food products against microorganisms during storage, as a flavoring in the production of chocolate and confectionery products, as an important ingredient in fragrance, and as a preservative in medical products. However, little is known of its effect on insects. The main aim of this study was to determine the influence of benzyl alcohol on the defense systems of the wax moth Galleria mellonella, i.e., its cuticular lipid composition and critical elements of its immune system. A gas chromatography/mass spectrometry (GC/MS) analysis found benzyl alcohol treatment to elicit significant quantitative and qualitative differences in cuticular free fatty acid (FFA) profiles. Our findings indicate that benzyl alcohol treatment increased the levels of HSP70 and HSP90 and decreased those of HSF1, histamine, and cysteinyl leukotriene. Benzyl alcohol application also increased dismutase level in the hemolymph and lowered those of catalase and 8-OHdG. The treatment also had negative effects on G. mellonella hemocytes and a Sf9 cell line in vitro: 48-h treatment resulted in morphological changes, with the remaining cells being clearly spindle-shaped with numerous granules. The high insecticidal activity of compound and its lack of toxicity towards vertebrates suggest it could be an effective insecticide.
Collapse
Affiliation(s)
- Michalina Kazek
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, ul. Poleczki 19, 02-822 Warszawa, Poland;
| | - Agata Kaczmarek
- Museum and Institute of Zoology, Polish Academy of Sciences, ul. Twarda 51/55, 00-818 Warszawa, Poland; (A.K.); (A.K.W.)
| | - Anna K. Wrońska
- Museum and Institute of Zoology, Polish Academy of Sciences, ul. Twarda 51/55, 00-818 Warszawa, Poland; (A.K.); (A.K.W.)
| | - Mieczysława I. Boguś
- Museum and Institute of Zoology, Polish Academy of Sciences, ul. Twarda 51/55, 00-818 Warszawa, Poland; (A.K.); (A.K.W.)
- BIOMIBO, ul. Strzygłowska 15, 04-872 Warszawa, Poland
| |
Collapse
|
10
|
Wang S, Miao S, Li Y, Wang J, Li C, Lu Y, Li B. Morphological and functional characterization of circulating hemocytes in Tribolium castaneum larvae. INSECT SCIENCE 2024. [PMID: 39361781 DOI: 10.1111/1744-7917.13455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 10/05/2024]
Abstract
Hemocytes are pivotal in the immune response of insects against invasive pathogens. However, our knowledge of hemocyte types and their specific function in Tribolium castaneum, an increasingly important Coleoptera model insect in various research fields, remains limited. Presently, a combination of morphological criteria and dye-staining properties were used to characterize hemocyte types from T. castaneum larvae, and 4 distinct types were identified: granulocytes, oenocytoids, plasmatocytes and prohemocytes. Following different immune challenges, the total hemocyte counts declined rapidly in the initial phase (at 2 h), then increased over time (at 4 and 6 h) and eventually returned to the naive state by 24 h post-injection. Notably, the morphology of granulocytes underwent dramatic changes, characterized by an expansion of the surface area and an increased production of pseudopods, and with the number of granulocytes rising significantly through mitotic division. Granulocytes and plasmatocytes, the main hemocyte types in T. castaneum larvae, can phagocytose bacteria or latex beads injected into the larval hemolymph in vivo. Furthermore, these hemocytes participate in the encapsulation and melanization processes in vitro, forming capsules to encapsulate and melanize nickel-nitrilotriacetic acid (Ni-NTA) beads. This study provides the first comprehensive characterization of circulating hemocytes in T. castaneum larvae, offering valuable insights into cell-mediated immunity in response to bacterial infection and the injection of latex beads. These results deepen our understanding of the cellular response mechanisms in T. castaneum larvae and lay a solid foundation for subsequent investigations of the involvement of T. castaneum hemocytes in combating pathogens.
Collapse
Affiliation(s)
- Suisui Wang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Shiyuan Miao
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- College of Environmental and Life Science, Murdoch University, Murdoch, WA, Australia
| | - Yusi Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jianhui Wang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chengjun Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yujie Lu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
11
|
Ratcliffe NA. Back to the future: Forgotten protocols for optimizing the isolation of arthropod haemocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105223. [PMID: 38960294 DOI: 10.1016/j.dci.2024.105223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Consideration is given to previous and more recent protocols for harvesting arthropod haemocytes from Galleria, Drosophila, mosquitoes, Limulus and crustaceans. The optimal harvesting of these cells is essential for meaningful studies of invertebrate immunity in vitro. The results of such experiments, however, have often been flawed due to a lack of understanding of the fragile nature of arthropod haemocytes on exposure to bacterial lipopolysaccharides, resulting in the aggregation and loss of cell types during haemolymph clotting. This article emphasizes that although there are similarities between mammalian neutrophils and arthropod haemocytes, the protocols required for the successful harvesting of these cells vary significantly. The various stages for the successful harvesting of arthropod haemocytes are described in detail and should provide invaluable advice to those requiring both high cell viability and recovery of the different cell types for subsequent experimentation.
Collapse
Affiliation(s)
- Norman A Ratcliffe
- Biology Institute, Universidade Federal Fluminense, Niterói, RJ, 24210-130, Brazil; Department of Biosciences, Swansea University, Singleton Park, Swansea, SA28PP, UK.
| |
Collapse
|
12
|
Brantley SE, Stouthamer CM, Kr P, Fischer ML, Hill J, Schlenke TA, Mortimer NT. Host JAK-STAT activity is a target of parasitoid wasp virulence strategies. PLoS Pathog 2024; 20:e1012349. [PMID: 38950076 PMCID: PMC11244843 DOI: 10.1371/journal.ppat.1012349] [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: 12/22/2023] [Revised: 07/12/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Innate immune responses that allow hosts to survive infection depend on the action of multiple conserved signaling pathways. Pathogens and parasites in turn have evolved virulence factors to target these immune signaling pathways in an attempt to overcome host immunity. Consequently, the interactions between host immune molecules and pathogen virulence factors play an important role in determining the outcome of an infection. The immune responses of Drosophila melanogaster provide a valuable model to understand immune signaling and host-pathogen interactions. Flies are commonly infected by parasitoid wasps and mount a coordinated cellular immune response following infection. This response is characterized by the production of specialized blood cells called lamellocytes that form a tight capsule around wasp eggs in the host hemocoel. The conserved JAK-STAT signaling pathway has been implicated in lamellocyte proliferation and is required for successful encapsulation of wasp eggs. Here we show that activity of Stat92E, the D. melanogaster STAT ortholog, is induced in immune tissues following parasitoid infection. Virulent wasp species are able to suppress Stat92E activity during infection, suggesting they target JAK-STAT pathway activation as a virulence strategy. Furthermore, two wasp species (Leptopilina guineaensis and Ganaspis xanthopoda) suppress phenotypes associated with a gain-of-function mutation in hopscotch, the D. melanogaster JAK ortholog, indicating that they inhibit the activity of the core signaling components of the JAK-STAT pathway. Our data suggest that parasitoid wasp virulence factors block JAK-STAT signaling to overcome fly immune defenses.
Collapse
Affiliation(s)
- Susanna E Brantley
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Corinne M Stouthamer
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Pooja Kr
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Mary L Fischer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Joshua Hill
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Todd A Schlenke
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Nathan T Mortimer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
13
|
Shin M, Chang E, Lee D, Kim N, Cho B, Cha N, Koranteng F, Song JJ, Shim J. Drosophila immune cells transport oxygen through PPO2 protein phase transition. Nature 2024; 631:350-359. [PMID: 38926577 PMCID: PMC11236712 DOI: 10.1038/s41586-024-07583-x] [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] [Received: 04/28/2022] [Accepted: 05/17/2024] [Indexed: 06/28/2024]
Abstract
Insect respiration has long been thought to be solely dependent on an elaborate tracheal system without assistance from the circulatory system or immune cells1,2. Here we describe that Drosophila crystal cells-myeloid-like immune cells called haemocytes-control respiration by oxygenating Prophenoloxidase 2 (PPO2) proteins. Crystal cells direct the movement of haemocytes between the trachea of the larval body wall and the circulation to collect oxygen. Aided by copper and a neutral pH, oxygen is trapped in the crystalline structures of PPO2 in crystal cells. Conversely, PPO2 crystals can be dissolved when carbonic anhydrase lowers the intracellular pH and then reassembled into crystals in cellulo by adhering to the trachea. Physiologically, larvae lacking crystal cells or PPO2, or those expressing a copper-binding mutant of PPO2, display hypoxic responses under normoxic conditions and are susceptible to hypoxia. These hypoxic phenotypes can be rescued by hyperoxia, expression of arthropod haemocyanin or prevention of larval burrowing activity to expose their respiratory organs. Thus, we propose that insect immune cells collaborate with the tracheal system to reserve and transport oxygen through the phase transition of PPO2 crystals, facilitating internal oxygen homeostasis in a process that is comparable to vertebrate respiration.
Collapse
Affiliation(s)
- Mingyu Shin
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Eunji Chang
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Daewon Lee
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Nayun Kim
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Bumsik Cho
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Nuri Cha
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Ferdinand Koranteng
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jiwon Shim
- Department of Life Science, College of Natural Science, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea.
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Maurya D, Rai G, Mandal D, Mondal BC. Transient caspase-mediated activation of caspase-activated DNase causes DNA damage required for phagocytic macrophage differentiation. Cell Rep 2024; 43:114251. [PMID: 38761374 PMCID: PMC7617294 DOI: 10.1016/j.celrep.2024.114251] [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: 12/05/2023] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
Phagocytic macrophages are crucial for innate immunity and tissue homeostasis. Most tissue-resident macrophages develop from embryonic precursors that populate every organ before birth to lifelong self-renew. However, the mechanisms for versatile macrophage differentiation remain unknown. Here, we use in vivo genetic and cell biological analysis of the Drosophila larval hematopoietic organ, the lymph gland that produces macrophages. We show that the developmentally regulated transient activation of caspase-activated DNase (CAD)-mediated DNA strand breaks in intermediate progenitors is essential for macrophage differentiation. Insulin receptor-mediated PI3K/Akt signaling regulates the apoptosis signal-regulating kinase 1 (Ask1)/c-Jun kinase (JNK) axis to control sublethal levels of caspase activation, causing DNA strand breaks during macrophage development. Furthermore, caspase activity is also required for embryonic-origin macrophage development and efficient phagocytosis. Our study provides insights into developmental signaling and CAD-mediated DNA strand breaks associated with multifunctional and heterogeneous macrophage differentiation.
Collapse
Affiliation(s)
- Deepak Maurya
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Gayatri Rai
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Debleena Mandal
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Bama Charan Mondal
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
15
|
Guo R, Ma G, Zhai X, Shi H, Wang J. Single-cell transcriptomic landscape of peripheral blood cells provides insights into adaptation of red-eared sliders (Trachemys scripta elegans). Integr Zool 2024; 19:468-479. [PMID: 37226359 DOI: 10.1111/1749-4877.12725] [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: 05/26/2023]
Abstract
Red-eared sliders (Trachemys scripta elegans), as one of the 100 most threatening aliens, have stronger immunity than the native species in response to environmental stress. Blood cells are an important component of immunity in the body. However, the blood cell researches of turtle are still in the traditional blood cell classification and morphological structure observation. Furthermore, turtle granulocytes cannot be accurately identified using traditional methods. Single-cell RNA sequencing techniques have been successfully implemented to study cells based on the mRNA expression patterns of each cell. The present study profiled the transcriptomes of peripheral blood cells in red-eared sliders to construct a single-cell transcriptional landscape of the different cell types and explored environmental adaptation mechanism from the perspective of hematology. All 14 transcriptionally distinct clusters (platelets, erythrocytes1, erythrocytes2, CSF1R monocytes, POF1B monocytes, neutrophils, GATA2high basophils, GATA2low basophils, CD4 T cells, CD7 T cells, B cells, ACKR4 cells, serotriflin cells, and ficolin cells) were identified in the peripheral blood cells of the red-eared sliders. In particular, a subtype of erythrocytes (erythrocytes1) that expressed immune signals was identified. Peripheral blood cells were grouped into three lineages: platelet, erythroid/lymphoid, and myeloid cell lineages. Furthermore, based on differentiation trajectory and up-regulated gene expression, ACKR4 cells were newly identified as lymphocytes, and serotriflin and ficolin cells as granulocytes. The single-cell transcriptional atlas of the peripheral blood cells in red-eared sliders provided in the present study will offer a comprehensive transcriptome reference for the exploration of physiological and pathological hematology in this species.
Collapse
Affiliation(s)
- Rui Guo
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Guangwei Ma
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Xiaofei Zhai
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Haitao Shi
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| | - Jichao Wang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, China
| |
Collapse
|
16
|
Kazek M, Chodáková L, Lehr K, Strych L, Nedbalová P, McMullen E, Bajgar A, Opekar S, Šimek P, Moos M, Doležal T. Glucose and trehalose metabolism through the cyclic pentose phosphate pathway shapes pathogen resistance and host protection in Drosophila. PLoS Biol 2024; 22:e3002299. [PMID: 38713712 PMCID: PMC11101078 DOI: 10.1371/journal.pbio.3002299] [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: 07/28/2023] [Revised: 05/17/2024] [Accepted: 04/12/2024] [Indexed: 05/09/2024] Open
Abstract
Activation of immune cells requires the remodeling of cell metabolism in order to support immune function. We study these metabolic changes through the infection of Drosophila larvae by parasitoid wasp. The parasitoid egg is neutralized by differentiating lamellocytes, which encapsulate the egg. A melanization cascade is initiated, producing toxic molecules to destroy the egg while the capsule also protects the host from the toxic reaction. We combined transcriptomics and metabolomics, including 13C-labeled glucose and trehalose tracing, as well as genetic manipulation of sugar metabolism to study changes in metabolism, specifically in Drosophila hemocytes. We found that hemocytes increase the expression of several carbohydrate transporters and accordingly uptake more sugar during infection. These carbohydrates are metabolized by increased glycolysis, associated with lactate production, and cyclic pentose phosphate pathway (PPP), in which glucose-6-phosphate is re-oxidized to maximize NADPH yield. Oxidative PPP is required for lamellocyte differentiation and resistance, as is systemic trehalose metabolism. In addition, fully differentiated lamellocytes use a cytoplasmic form of trehalase to cleave trehalose to glucose and fuel cyclic PPP. Intracellular trehalose metabolism is not required for lamellocyte differentiation, but its down-regulation elevates levels of reactive oxygen species, associated with increased resistance and reduced fitness. Our results suggest that sugar metabolism, and specifically cyclic PPP, within immune cells is important not only to fight infection but also to protect the host from its own immune response and for ensuring fitness of the survivor.
Collapse
Affiliation(s)
- Michalina Kazek
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lenka Chodáková
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Katharina Lehr
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lukáš Strych
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Pavla Nedbalová
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Ellen McMullen
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Adam Bajgar
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Stanislav Opekar
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Petr Šimek
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Martin Moos
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Tomáš Doležal
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| |
Collapse
|
17
|
Leitão AB, Arunkumar R, Day JP, Hanna N, Devi A, Hayes MP, Jiggins FM. Recognition of nonself is necessary to activate Drosophila's immune response against an insect parasite. BMC Biol 2024; 22:89. [PMID: 38644510 PMCID: PMC11034056 DOI: 10.1186/s12915-024-01886-1] [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: 07/27/2023] [Accepted: 04/11/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Innate immune responses can be activated by pathogen-associated molecular patterns (PAMPs), danger signals released by damaged tissues, or the absence of self-molecules that inhibit immunity. As PAMPs are typically conserved across broad groups of pathogens but absent from the host, it is unclear whether they allow hosts to recognize parasites that are phylogenetically similar to themselves, such as parasitoid wasps infecting insects. RESULTS Parasitoids must penetrate the cuticle of Drosophila larvae to inject their eggs. In line with previous results, we found that the danger signal of wounding triggers the differentiation of specialized immune cells called lamellocytes. However, using oil droplets to mimic infection by a parasitoid wasp egg, we found that this does not activate the melanization response. This aspect of the immune response also requires exposure to parasite molecules. The unidentified factor enhances the transcriptional response in hemocytes and induces a specific response in the fat body. CONCLUSIONS We conclude that a combination of danger signals and the recognition of nonself molecules is required to activate Drosophila's immune response against parasitic insects.
Collapse
Affiliation(s)
- Alexandre B Leitão
- Department of Genetics, University of Cambridge, Cambridge, UK.
- Champalimaud Foundation, Lisbon, Portugal.
| | | | - Jonathan P Day
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Nancy Hanna
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Aarathi Devi
- Department of Genetics, University of Cambridge, Cambridge, UK
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Matthew P Hayes
- Department of Zoology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
18
|
Baassiri A, Ghais A, Kurdi A, Rahal E, Nasr R, Shirinian M. The molecular signature of BCR::ABLP210 and BCR::ABLT315I in a Drosophila melanogaster chronic myeloid leukemia model. iScience 2024; 27:109538. [PMID: 38585663 PMCID: PMC10995885 DOI: 10.1016/j.isci.2024.109538] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal hematopoietic stem cell disorder resulting from a balanced translocation leading to BCR::ABL1 oncogene with increased tyrosine kinase activity. Despite the advancements in the development of tyrosine kinase inhibitors (TKIs), the T315I gatekeeper point mutation in the BCR::ABL1 gene remains a challenge. We have previously reported in a Drosophila CML model an increased hemocyte count and disruption in sessile hemocyte patterns upon expression of BCR::ABL1p210 and BCR::ABL1T315I in the hemolymph. In this study, we performed RNA sequencing to determine if there is a distinct gene expression that distinguishes BCR::ABL1p210 and BCR::ABL1T315I. We identified six genes that were consistently upregulated in the fly CML model and validated in adult and pediatric CML patients and in a mouse cell line expressing BCR::ABL1T315I. This study provides a comprehensive analysis of gene signatures in BCR::ABL1p210 and BCR::ABL1T315I, laying the groundwork for targeted investigations into the role of these genes in CML pathogenesis.
Collapse
Affiliation(s)
- Amro Baassiri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Ghais
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Abdallah Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Elias Rahal
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Margret Shirinian
- Department of Experimental Pathology and Immunology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
19
|
Sinenko SA. Molecular Mechanisms of Drosophila Hematopoiesis. Acta Naturae 2024; 16:4-21. [PMID: 39188265 PMCID: PMC11345091 DOI: 10.32607/actanaturae.27410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/31/2024] [Indexed: 08/28/2024] Open
Abstract
As a model organism, the fruit fly (Drosophila melanogaster) has assumed a leading position in modern biological research. The Drosophila genetic system has a number of advantages making it a key model in investigating the molecular mechanisms of metazoan developmental processes. Over the past two decades, significant progress has been made in understanding the molecular mechanisms regulating Drosophila hematopoiesis. This review discusses the major advances in investigating the molecular mechanisms involved in maintaining the population of multipotent progenitor cells and their differentiation into mature hemocytes in the hematopoietic organ of the Drosophila larva. The use of the Drosophila hematopoietic organ as a model system for hematopoiesis has allowed to characterize the complex interactions between signaling pathways and transcription factors in regulating the maintenance and differentiation of progenitor cells through the signals from the hematopoietic niche, autocrine and paracrine signals, and the signals emanated by differentiated cells.
Collapse
Affiliation(s)
- S. A. Sinenko
- Institute of Cytology Russian Academy of Sciences, St. Petersburg, 194064 Russian Federation
| |
Collapse
|
20
|
Krejčová G, Danielová A, Sehadová H, Dyčka F, Kubásek J, Moos M, Bajgar A. Macrophages play a nutritive role in post-metamorphic maturation in Drosophila. Development 2024; 151:dev202492. [PMID: 38456486 DOI: 10.1242/dev.202492] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
In the body of multicellular organisms, macrophages play an indispensable role in maintaining tissue homeostasis by removing old, apoptotic and damaged cells. In addition, macrophages allow significant remodeling of body plans during embryonic morphogenesis, regeneration and metamorphosis. Although the huge amount of organic matter that must be removed during these processes represents a potential source of nutrients, their further use by the organism has not yet been addressed. Here, we document that, during metamorphosis, Drosophila larval adipose tissue is infiltrated by macrophages, which remove dying adipocytes by efferocytosis and engulf leaking RNA-protein granules and lipids. Consequently, the infiltrating macrophages transiently adopt the adipocyte-like metabolic profile to convert remnants of dying adipocytes to lipoproteins and storage peptides that nutritionally support post-metamorphic development. This process is fundamental for the full maturation of ovaries and the achievement of early fecundity of individuals. Whether macrophages play an analogous role in other situations of apoptotic cell removal remains to be elucidated.
Collapse
Affiliation(s)
- Gabriela Krejčová
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Adéla Danielová
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Hana Sehadová
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| | - Filip Dyčka
- Department of Chemistry, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Jiří Kubásek
- Department of Experimental Plant Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
| | - Martin Moos
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| | - Adam Bajgar
- Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia, Ceske Budejovice, 37005, Czech Republic
- Institute of Entomology , Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, 37005, Czech Republic
| |
Collapse
|
21
|
Deichsel S, Gahr BM, Mastel H, Preiss A, Nagel AC. Numerous Serine/Threonine Kinases Affect Blood Cell Homeostasis in Drosophila melanogaster. Cells 2024; 13:576. [PMID: 38607015 PMCID: PMC11011202 DOI: 10.3390/cells13070576] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Blood cells in Drosophila serve primarily innate immune responses. Various stressors influence blood cell homeostasis regarding both numbers and the proportion of blood cell types. The principle molecular mechanisms governing hematopoiesis are conserved amongst species and involve major signaling pathways like Notch, Toll, JNK, JAK/Stat or RTK. Albeit signaling pathways generally rely on the activity of protein kinases, their specific contribution to hematopoiesis remains understudied. Here, we assess the role of Serine/Threonine kinases with the potential to phosphorylate the transcription factor Su(H) in crystal cell homeostasis. Su(H) is central to Notch signal transduction, and its inhibition by phosphorylation impedes crystal cell formation. Overall, nearly twenty percent of all Drosophila Serine/Threonine kinases were studied in two assays, global and hemocyte-specific overexpression and downregulation, respectively. Unexpectedly, the majority of kinases influenced crystal cell numbers, albeit only a few were related to hematopoiesis so far. Four kinases appeared essential for crystal cell formation, whereas most kinases restrained crystal cell development. This group comprises all kinase classes, indicative of the complex regulatory network underlying blood cell homeostasis. The rather indiscriminative response we observed opens the possibility that blood cells measure their overall phospho-status as a proxy for stress-signals, and activate an adaptive immune response accordingly.
Collapse
Affiliation(s)
- Sebastian Deichsel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Bernd M. Gahr
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Helena Mastel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Anette Preiss
- Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Anja C. Nagel
- Department of Molecular Genetics, Institute of Biology, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
22
|
Cui C, Tang X, Xing J, Sheng X, Chi H, Zhan W. Single-cell RNA-seq revealed heterogeneous responses and functional differentiation of hemocytes against white spot syndrome virus infection in Litopenaeus vannamei. J Virol 2024; 98:e0180523. [PMID: 38323810 PMCID: PMC10949519 DOI: 10.1128/jvi.01805-23] [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: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Shrimp hemocytes are the vital immune cells participating in innate immune response to defend against viruses. However, the lack of specific molecular markers for shrimp hemocyte hindered the insightful understanding of their functional clusters and differential roles in combating microbial infections. In this study, we used single-cell RNA sequencing to map the transcriptomic landscape of hemocytes from the white spot syndrome virus (WSSV)-infected Litopenaeus vannamei and conjointly analyzed with our previous published single-cell RNA sequencing technology data from the healthy hemocytes. A total of 16 transcriptionally distinct cell clusters were identified, which occupied different proportions in healthy and WSSV-infected hemocytes and exerted differential roles in antiviral immune response. Following mapping of the sequencing data to the WSSV genome, we found that all types of hemocytes could be invaded by WSSV virions, especially the cluster 8, which showed the highest transcriptional levels of WSSV genes and exhibited a cell type-specific antiviral response to the viral infection. Further evaluation of the cell clusters revealed the delicate dynamic balance between hemocyte immune response and viral infestation. Unsupervised pseudo-time analysis of hemocytes showed that the hemocytes in immune-resting state could be significantly activated upon WSSV infection and then functionally differentiated to different hemocyte subsets. Collectively, our results revealed the differential responses of shrimp hemocytes and the process of immune-functional differentiation post-WSSV infection, providing essential resource for the systematic insight into the synergistic immune response mechanism against viral infection among hemocyte subtypes. IMPORTANCE Current knowledge of shrimp hemocyte classification mainly comes from morphology, which hinder in-depth characterization of cell lineage development, functional differentiation, and different immune response of hemocyte types during pathogenic infections. Here, single-cell RNA sequencing was used for mapping hemocytes during white spot syndrome virus (WSSV) infection in Litopenaeus vannamei, identifying 16 cell clusters and evaluating their potential antiviral functional characteristics. We have described the dynamic balance between viral infestation and hemocyte immunity. And the functional differentiation of hemocytes under WSSV stimulation was further characterized. Our results provided a comprehensive transcriptional landscape and revealed the heterogeneous immune response in shrimp hemocytes during WSSV infection.
Collapse
Affiliation(s)
- Chuang Cui
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
23
|
Ratcliffe NA, Mello CB, Castro HC, Dyson P, Figueiredo M. Immune Reactions of Vector Insects to Parasites and Pathogens. Microorganisms 2024; 12:568. [PMID: 38543619 PMCID: PMC10974449 DOI: 10.3390/microorganisms12030568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 11/12/2024] Open
Abstract
This overview initially describes insect immune reactions and then brings together present knowledge of the interactions of vector insects with their invading parasites and pathogens. It is a way of introducing this Special Issue with subsequent papers presenting the latest details of these interactions in each particular group of vectors. Hopefully, this paper will fill a void in the literature since brief descriptions of vector immunity have now been brought together in one publication and could form a starting point for those interested and new to this important area. Descriptions are given on the immune reactions of mosquitoes, blackflies, sandflies, tsetse flies, lice, fleas and triatomine bugs. Cellular and humoral defences are described separately but emphasis is made on the co-operation of these processes in the completed immune response. The paper also emphasises the need for great care in extracting haemocytes for subsequent study as appreciation of their fragile nature is often overlooked with the non-sterile media, smearing techniques and excessive centrifugation sometimes used. The potential vital role of eicosanoids in the instigation of many of the immune reactions described is also discussed. Finally, the priming of the immune system, mainly in mosquitoes, is considered and one possible mechanism is presented.
Collapse
Affiliation(s)
- Norman Arthur Ratcliffe
- Department of Biosciences, Swansea University, Singleton Park, Swansea SA28PP, UK
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Cicero Brasileiro Mello
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Helena Carla Castro
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Paul Dyson
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| | - Marcela Figueiredo
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| |
Collapse
|
24
|
Rolandelli A, Laukaitis-Yousey HJ, Bogale HN, Singh N, Samaddar S, O'Neal AJ, Ferraz CR, Butnaru M, Mameli E, Xia B, Mendes MT, Butler LR, Marnin L, Cabrera Paz FE, Valencia LM, Rana VS, Skerry C, Pal U, Mohr SE, Perrimon N, Serre D, Pedra JHF. Tick hemocytes have a pleiotropic role in microbial infection and arthropod fitness. Nat Commun 2024; 15:2117. [PMID: 38459063 PMCID: PMC10923820 DOI: 10.1038/s41467-024-46494-3] [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: 10/20/2023] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
Uncovering the complexity of systems in non-model organisms is critical for understanding arthropod immunology. Prior efforts have mostly focused on Dipteran insects, which only account for a subset of existing arthropod species in nature. Here we use and develop advanced techniques to describe immune cells (hemocytes) from the clinically relevant tick Ixodes scapularis at a single-cell resolution. We observe molecular alterations in hemocytes upon feeding and infection with either the Lyme disease spirochete Borrelia burgdorferi or the rickettsial agent Anaplasma phagocytophilum. We reveal hemocyte clusters exhibiting defined signatures related to immunity, metabolism, and proliferation. Depletion of phagocytic hemocytes affects hemocytin and astakine levels, two I. scapularis hemocyte markers, impacting blood-feeding, molting behavior, and bacterial acquisition. Mechanistically, astakine alters hemocyte proliferation, whereas hemocytin affects the c-Jun N-terminal kinase (JNK) signaling pathway in I. scapularis. Altogether, we discover a role for tick hemocytes in immunophysiology and provide a valuable resource for comparative biology in arthropods.
Collapse
Affiliation(s)
- Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanna J Laukaitis-Yousey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Haikel N Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Rancho BioSciences, San Diego, CA, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University; Knowledge Corridor, Gandhinagar, Gujarat, India
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Camila R Ferraz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, USA
| | - Baolong Xia
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - M Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luisa M Valencia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vipin S Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David Serre
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Saha B, McNinch CM, Lu S, Ho MCW, De Carvalho SS, Barillas-Mury C. In-depth transcriptomic analysis of Anopheles gambiae hemocytes uncovers novel genes and the oenocytoid developmental lineage. BMC Genomics 2024; 25:80. [PMID: 38243165 PMCID: PMC10799387 DOI: 10.1186/s12864-024-09986-6] [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: 09/28/2023] [Accepted: 01/07/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Hemocytes are immune cells that patrol the mosquito hemocoel and mediate critical cellular defense responses against pathogens. However, despite their importance, a comprehensive transcriptome of these cells was lacking because they constitute a very small fraction of the total cells in the insect, limiting the study of hemocyte differentiation and immune function. RESULTS In this study, an in-depth hemocyte transcriptome was built by extensive bulk RNA sequencing and assembly of hemocyte RNAs from adult A. gambiae female mosquitoes, based on approximately 2.4 billion short Illumina and about 9.4 million long PacBio high-quality reads that mapped to the A. gambiae PEST genome (P4.14 version). A total of 34,939 transcripts were annotated including 4,020 transcripts from novel genes and 20,008 novel isoforms that result from extensive differential splicing of transcripts from previously annotated genes. Most hemocyte transcripts identified (89.8%) are protein-coding while 10.2% are non-coding RNAs. The number of transcripts identified in the novel hemocyte transcriptome is twice the number in the current annotation of the A. gambiae genome (P4.14 version). Furthermore, we were able to refine the analysis of a previously published single-cell transcriptome (scRNAseq) data set by using the novel hemocyte transcriptome as a reference to re-define the hemocyte clusters and determine the path of hemocyte differentiation. Unsupervised pseudo-temporal ordering using the Tools for Single Cell Analysis software uncovered a novel putative prohemocyte precursor cell type that gives rise to prohemocytes. Pseudo-temporal ordering with the Monocle 3 software, which analyses changes in gene expression during dynamic biological processes, determined that oenocytoids derive from prohemocytes, a cell population that also gives rise to the granulocyte lineage. CONCLUSION A high number of mRNA splice variants are expressed in hemocytes, and they may account for the plasticity required to mount efficient responses to many different pathogens. This study highlights the importance of a comprehensive set of reference transcripts to perform robust single-cell transcriptomic data analysis of cells present in low abundance. The detailed annotation of the hemocyte transcriptome will uncover new facets of hemocyte development and function in adult dipterans and is a valuable community resource for future studies on mosquito cellular immunity.
Collapse
Affiliation(s)
- Banhisikha Saha
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Colton M McNinch
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, 20892, Bethesda, MD, USA
| | - Stephen Lu
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Margaret C W Ho
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, 20892, Bethesda, MD, USA
| | - Stephanie Serafim De Carvalho
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA.
| |
Collapse
|
26
|
Kloc M, Halasa M, Kubiak JZ, Ghobrial RM. Invertebrate Immunity, Natural Transplantation Immunity, Somatic and Germ Cell Parasitism, and Transposon Defense. Int J Mol Sci 2024; 25:1072. [PMID: 38256145 PMCID: PMC10815962 DOI: 10.3390/ijms25021072] [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: 12/06/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
While the vertebrate immune system consists of innate and adaptive branches, invertebrates only have innate immunity. This feature makes them an ideal model system for studying the cellular and molecular mechanisms of innate immunity sensu stricto without reciprocal interferences from adaptive immunity. Although invertebrate immunity is evolutionarily older and a precursor of vertebrate immunity, it is far from simple. Despite lacking lymphocytes and functional immunoglobulin, the invertebrate immune system has many sophisticated mechanisms and features, such as long-term immune memory, which, for decades, have been exclusively attributed to adaptive immunity. In this review, we describe the cellular and molecular aspects of invertebrate immunity, including the epigenetic foundation of innate memory, the transgenerational inheritance of immunity, genetic immunity against invading transposons, the mechanisms of self-recognition, natural transplantation, and germ/somatic cell parasitism.
Collapse
Affiliation(s)
- Malgorzata Kloc
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA; (M.H.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Marta Halasa
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA; (M.H.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Jacek Z. Kubiak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine-National Research Institute (WIM-PIB), Szaserow 128, 04-141 Warsaw, Poland;
- Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, CNRS, UMR 6290, 35043 Rennes, France
| | - Rafik M. Ghobrial
- Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA; (M.H.); (R.M.G.)
- Department of Surgery, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
27
|
Brooks EC, Zeidler MP, Ong ACM, Evans IR. Macrophage subpopulation identity in Drosophila is modulated by apoptotic cell clearance and related signalling pathways. Front Immunol 2024; 14:1310117. [PMID: 38283366 PMCID: PMC10811221 DOI: 10.3389/fimmu.2023.1310117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
In Drosophila blood, plasmatocytes of the haemocyte lineage represent the functional equivalent of vertebrate macrophages and have become an established in vivo model with which to study macrophage function and behaviour. However, the use of plasmatocytes as a macrophage model has been limited by a historical perspective that plasmatocytes represent a homogenous population of cells, in contrast to the high levels of heterogeneity of vertebrate macrophages. Recently, a number of groups have reported transcriptomic approaches which suggest the existence of plasmatocyte heterogeneity, while we identified enhancer elements that identify subpopulations of plasmatocytes which exhibit potentially pro-inflammatory behaviours, suggesting conservation of plasmatocyte heterogeneity in Drosophila. These plasmatocyte subpopulations exhibit enhanced responses to wounds and decreased rates of efferocytosis when compared to the overall plasmatocyte population. Interestingly, increasing the phagocytic requirement placed upon plasmatocytes is sufficient to decrease the size of these plasmatocyte subpopulations in the embryo. However, the mechanistic basis for this response was unclear. Here, we examine how plasmatocyte subpopulations are modulated by apoptotic cell clearance (efferocytosis) demands and associated signalling pathways. We show that loss of the phosphatidylserine receptor Simu prevents an increased phagocytic burden from modulating specific subpopulation cells, while blocking other apoptotic cell receptors revealed no such rescue. This suggests that Simu-dependent efferocytosis is specifically involved in determining fate of particular subpopulations. Supportive of our original finding, mutations in amo (the Drosophila homolog of PKD2), a calcium-permeable channel which operates downstream of Simu, phenocopy simu mutants. Furthermore, we show that Amo is involved in the acidification of the apoptotic cell-containing phagosomes, suggesting that this reduction in pH may be associated with macrophage reprogramming. Additionally, our results also identify Ecdysone receptor signalling, a pathway related to control of cell death during developmental transitions, as a controller of plasmatocyte subpopulation identity. Overall, these results identify fundamental pathways involved in the specification of plasmatocyte subpopulations and so further validate Drosophila plasmatocytes as a heterogeneous population of macrophage-like cells within this important developmental and immune model.
Collapse
Affiliation(s)
- Elliot C. Brooks
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Martin P. Zeidler
- School of Biosciences and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Albert C. M. Ong
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Iwan R. Evans
- School of Medicine and Population Health and the Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
28
|
Adegoke A, Ribeiro JMC, Smith R, Karim S. Tick innate immune responses to hematophagy and Ehrlichia infection at single-cell resolution. Front Immunol 2024; 14:1305976. [PMID: 38274813 PMCID: PMC10808623 DOI: 10.3389/fimmu.2023.1305976] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Ticks rely on robust cellular and humoral responses to control microbial infection. However, several aspects of the tick's innate immune system remain uncharacterized, most notably that of the immune cells (called hemocytes), which are known to play a significant role in cellular and humoral responses. Despite the importance of hemocytes in regulating microbial infection, our understanding of their basic biology and molecular mechanisms remains limited. Therefore, we believe that a more detailed understanding of the role of hemocytes in the interactions between ticks and tick-borne microbes is crucial to illuminating their function in vector competence and to help identify novel targets for developing new strategies to block tick-borne pathogen transmission. Methods This study examined hemocytes from the lone star tick (Amblyomma americanum) at the transcriptomic level using the 10X genomics single-cell RNA sequencing platform to analyze hemocyte populations from unfed, partially blood-fed, and Ehrlichia chaffeensis-infected ticks. The functional role of differentially expressed hemocyte markers in hemocyte proliferation and Ehrlichia dissemination was determined using an RNA interference approach. Results and discussion Our data exhibit the identification of fourteen distinct hemocyte populations. Our results uncover seven distinct lineages present in uninfected and Ehrlichia-infected hemocyte clusters. The functional characterization of hemocytin, cystatin, fibronectin, and lipocalin demonstrate their role in hemocyte population changes, proliferation, and Ehrlichia dissemination. Conclusion Our results uncover the tick immune responses to Ehrlichia infection and hematophagy at a single-cell resolution. This work opens a new field of tick innate immunobiology to understand the role of hemocytes, particularly in response to prolonged blood-feeding (hematophagy), and tick-microbial interactions.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
29
|
Hersperger F, Meyring T, Weber P, Chhatbar C, Monaco G, Dionne MS, Paeschke K, Prinz M, Groß O, Classen AK, Kierdorf K. DNA damage signaling in Drosophila macrophages modulates systemic cytokine levels in response to oxidative stress. eLife 2024; 12:RP86700. [PMID: 38189792 PMCID: PMC10945508 DOI: 10.7554/elife.86700] [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: 01/09/2024] Open
Abstract
Environmental factors, infection, or injury can cause oxidative stress in diverse tissues and loss of tissue homeostasis. Effective stress response cascades, conserved from invertebrates to mammals, ensure reestablishment of homeostasis and tissue repair. Hemocytes, the Drosophila blood-like cells, rapidly respond to oxidative stress by immune activation. However, the precise signals how they sense oxidative stress and integrate these signals to modulate and balance the response to oxidative stress in the adult fly are ill-defined. Furthermore, hemocyte diversification was not explored yet on oxidative stress. Here, we employed high-throughput single nuclei RNA-sequencing to explore hemocytes and other cell types, such as fat body, during oxidative stress in the adult fly. We identified distinct cellular responder states in plasmatocytes, the Drosophila macrophages, associated with immune response and metabolic activation upon oxidative stress. We further define oxidative stress-induced DNA damage signaling as a key sensor and a rate-limiting step in immune-activated plasmatocytes controlling JNK-mediated release of the pro-inflammatory cytokine unpaired-3. We subsequently tested the role of this specific immune activated cell stage during oxidative stress and found that inhibition of DNA damage signaling in plasmatocytes, as well as JNK or upd3 overactivation, result in a higher susceptibility to oxidative stress. Our findings uncover that a balanced composition and response of hemocyte subclusters is essential for the survival of adult Drosophila on oxidative stress by regulating systemic cytokine levels and cross-talk to other organs, such as the fat body, to control energy mobilization.
Collapse
Affiliation(s)
- Fabian Hersperger
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
| | - Tim Meyring
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Pia Weber
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Chintan Chhatbar
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
| | - Gianni Monaco
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of FreiburgFreiburgGermany
| | - Marc S Dionne
- MRC Centre for Molecular Bacteriology and Infection, Imperial College LondonLondonUnited Kingdom
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Katrin Paeschke
- Department of Oncology, Haematology and Rheumatology, University Hospital BonnBonnGermany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital BonnBonnGermany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- Signalling Research Centres BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- Signalling Research Centres BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, Faculty of Biology, University of FreiburgFreiburgGermany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, Medical Center, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of FreiburgFreiburgGermany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| |
Collapse
|
30
|
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
|
31
|
Sun C, Shao Y, Iqbal J. Insect Insights at the Single-Cell Level: Technologies and Applications. Cells 2023; 13:91. [PMID: 38201295 PMCID: PMC10777908 DOI: 10.3390/cells13010091] [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: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Single-cell techniques are a promising way to unravel the complexity and heterogeneity of transcripts at the cellular level and to reveal the composition of different cell types and functions in a tissue or organ. In recent years, advances in single-cell RNA sequencing (scRNA-seq) have further changed our view of biological systems. The application of scRNA-seq in insects enables the comprehensive characterization of both common and rare cell types and cell states, the discovery of new cell types, and revealing how cell types relate to each other. The recent application of scRNA-seq techniques to insect tissues has led to a number of exciting discoveries. Here we provide an overview of scRNA-seq and its application in insect research, focusing on biological applications, current challenges, and future opportunities to make new discoveries with scRNA-seq in insects.
Collapse
Affiliation(s)
- Chao Sun
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Yongqi Shao
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junaid Iqbal
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
32
|
Cinege G, Magyar LB, Kovács H, Varga V, Bodai L, Zsindely N, Nagy G, Hegedűs Z, Hultmark D, Andó I. Distinctive features of Zaprionus indianus hemocyte differentiation and function revealed by transcriptomic analysis. Front Immunol 2023; 14:1322381. [PMID: 38187383 PMCID: PMC10768004 DOI: 10.3389/fimmu.2023.1322381] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Background Insects have specialized cell types that participate in the elimination of parasites, for instance, the lamellocytes of the broadly studied species Drosophila melanogaster. Other drosophilids, such as Drosophila ananassae and the invasive Zaprionus indianus, have multinucleated giant hemocytes, a syncytium of blood cells that participate in the encapsulation of the eggs or larvae of parasitoid wasps. These cells can be formed by the fusion of hemocytes in circulation or originate from the lymph gland. Their ultrastructure highly resembles that of the mammalian megakaryocytes. Methods Morphological, protein expressional, and functional features of blood cells were revealed using epifluorescence and confocal microscopy. The respective hemocyte subpopulations were identified using monoclonal antibodies in indirect immunofluorescence assays. Fluorescein isothiocyanate (FITC)-labeled Escherichia coli bacteria were used in phagocytosis tests. Gene expression analysis was performed following mRNA sequencing of blood cells. Results D. ananassae and Z. indianus encapsulate foreign particles with the involvement of multinucleated giant hemocytes and mount a highly efficient immune response against parasitoid wasps. Morphological, protein expressional, and functional assays of Z. indianus blood cells suggested that these cells could be derived from large plasmatocytes, a unique cell type developing specifically after parasitoid wasp infection. Transcriptomic analysis of blood cells, isolated from naïve and wasp-infected Z. indianus larvae, revealed several differentially expressed genes involved in signal transduction, cell movements, encapsulation of foreign targets, energy production, and melanization, suggesting their role in the anti-parasitoid response. A large number of genes that encode proteins associated with coagulation and wound healing, such as phenoloxidase activity factor-like proteins, fibrinogen-related proteins, lectins, and proteins involved in the differentiation and function of platelets, were constitutively expressed. The remarkable ultrastructural similarities between giant hemocytes and mammalian megakaryocytes, and presence of platelets, and giant cell-derived anucleated fragments at wound sites hint at the involvement of this cell subpopulation in wound healing processes, in addition to participation in the encapsulation reaction. Conclusion Our observations provide insights into the broad repertoire of blood cell functions required for efficient defense reactions to maintain the homeostasis of the organism. The analysis of the differentiation and function of multinucleated giant hemocytes gives an insight into the diversification of the immune mechanisms.
Collapse
Affiliation(s)
- Gyöngyi Cinege
- Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Lilla B. Magyar
- Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Henrietta Kovács
- Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Viktória Varga
- Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Nóra Zsindely
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Gábor Nagy
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Zoltán Hegedűs
- Laboratory of Bioinformatics, HUN-REN Biological Research Centre, Szeged, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Dan Hultmark
- Department of Molecular Biology, Umea University, Umea, Sweden
| | - István Andó
- Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| |
Collapse
|
33
|
Vincow ES, Thomas RE, Milstein G, Pareek G, Bammler T, MacDonald J, Pallanck L. Glucocerebrosidase deficiency leads to neuropathology via cellular immune activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571406. [PMID: 38168223 PMCID: PMC10760128 DOI: 10.1101/2023.12.13.571406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Mutations in GBA (glucosylceramidase beta), which encodes the lysosomal enzyme glucocerebrosidase (GCase), are the strongest genetic risk factor for the neurodegenerative disorders Parkinson's disease (PD) and Lewy body dementia. Recent work has suggested that neuroinflammation may be an important factor in the risk conferred by GBA mutations. We therefore systematically tested the contributions of immune-related genes to neuropathology in a Drosophila model of GCase deficiency. We identified target immune factors via RNA-Seq and proteomics on heads from GCase-deficient flies, which revealed both increased abundance of humoral factors and increased macrophage activation. We then manipulated the identified immune factors and measured their effect on head protein aggregates, a hallmark of neurodegenerative disease. Genetic ablation of humoral (secreted) immune factors did not suppress the development of protein aggregation. By contrast, re-expressing Gba1b in activated macrophages suppressed head protein aggregation in Gba1b mutants and rescued their lifespan and behavioral deficits. Moreover, reducing the GCase substrate glucosylceramide in activated macrophages also ameliorated Gba1b mutant phenotypes. Taken together, our findings show that glucosylceramide accumulation due to GCase deficiency leads to macrophage activation, which in turn promotes the development of neuropathology.
Collapse
Affiliation(s)
- Evelyn S. Vincow
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Ruth E. Thomas
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Gillian Milstein
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Gautam Pareek
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Leo Pallanck
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
34
|
Dolezal T. How to eliminate pathogen without killing oneself? Immunometabolism of encapsulation and melanization in Drosophila. Front Immunol 2023; 14:1330312. [PMID: 38124757 PMCID: PMC10730662 DOI: 10.3389/fimmu.2023.1330312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Cellular encapsulation associated with melanization is a crucial component of the immune response in insects, particularly against larger pathogens. The infection of a Drosophila larva by parasitoid wasps, like Leptopilina boulardi, is the most extensively studied example. In this case, the encapsulation and melanization of the parasitoid embryo is linked to the activation of plasmatocytes that attach to the surface of the parasitoid. Additionally, the differentiation of lamellocytes that encapsulate the parasitoid, along with crystal cells, is accountable for the melanization process. Encapsulation and melanization lead to the production of toxic molecules that are concentrated in the capsule around the parasitoid and, at the same time, protect the host from this toxic immune response. Thus, cellular encapsulation and melanization represent primarily a metabolic process involving the metabolism of immune cell activation and differentiation, the production of toxic radicals, but also the production of melanin and antioxidants. As such, it has significant implications for host physiology and systemic metabolism. Proper regulation of metabolism within immune cells, as well as at the level of the entire organism, is therefore essential for an efficient immune response and also impacts the health and overall fitness of the organism that survives. The purpose of this "perspective" article is to map what we know about the metabolism of this type of immune response, place it in the context of possible implications for host physiology, and highlight open questions related to the metabolism of this important insect immune response.
Collapse
Affiliation(s)
- Tomas Dolezal
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| |
Collapse
|
35
|
Yoon SH, Cho B, Lee D, Kim H, Shim J, Nam JW. Molecular traces of Drosophila hemocytes reveal transcriptomic conservation with vertebrate myeloid cells. PLoS Genet 2023; 19:e1011077. [PMID: 38113249 PMCID: PMC10763942 DOI: 10.1371/journal.pgen.1011077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 01/03/2024] [Accepted: 11/21/2023] [Indexed: 12/21/2023] Open
Abstract
Drosophila hemocytes serve as the primary defense system against harmful threats, allowing the animals to thrive. Hemocytes are often compared to vertebrate innate immune system cells due to the observed functional similarities between the two. However, the similarities have primarily been established based on a limited number of genes and their functional homologies. Thus, a systematic analysis using transcriptomic data could offer novel insights into Drosophila hemocyte function and provide new perspectives on the evolution of the immune system. Here, we performed cross-species comparative analyses using single-cell RNA sequencing data from Drosophila and vertebrate immune cells. We found several conserved markers for the cluster of differentiation (CD) genes in Drosophila hemocytes and validated the role of CG8501 (CD59) in phagocytosis by plasmatocytes, which function much like macrophages in vertebrates. By comparing whole transcriptome profiles in both supervised and unsupervised analyses, we showed that Drosophila hemocytes are largely homologous to vertebrate myeloid cells, especially plasmatocytes to monocytes/macrophages and prohemocyte 1 (PH1) to hematopoietic stem cells. Furthermore, a small subset of prohemocytes with hematopoietic potential displayed homology with hematopoietic progenitor populations in vertebrates. Overall, our results provide a deeper understanding of molecular conservation in the Drosophila immune system.
Collapse
Affiliation(s)
- Sang-Ho Yoon
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Bio-BigData Research Center, Hanyang University, Seoul, Republic of Korea
| | - Bumsik Cho
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Daewon Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Hanji Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jiwon Shim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Bio-BigData Research Center, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
| | - Jin-Wu Nam
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Bio-BigData Research Center, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
36
|
Bao Y, Shen G, Guo Y, Wang Q, Fan X, Li W. Effects of the tumor necrosis factor on hemocyte proliferation and bacterial infection in Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2023; 143:109175. [PMID: 37890735 DOI: 10.1016/j.fsi.2023.109175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023]
Abstract
Tumor necrosis factor (TNF) is an important cytokine that can regulate a variety of cellular responses by binding tumor necrosis factor receptor (TNFR). We studied whether the TNF of Eriocheir sinensis can regulate hemocyte proliferation. The results showed that the EsTNF and EsTNFR were constitutively expressed in all tested tissues, including the heart, hepatopancreas, muscles, gills, stomachs, intestines, and hemocytes. We found that low levels of EsTNF and EsTNFR transcripts were present in hemocytes. The gene expression levels were significantly increased in the hemocytes after being stimulated by Staphylococcus aureus or Vibrio parahaemolyticus. We also found some genes related to cell proliferation were expressed at a higher level in pulsing rTNF-stimulated hemocytes compared with the control group. We also knocked down the EsTNFR gene with RNAi technology. The results showed that the expression level of these genes related to cell proliferation was significantly down-regulated compared with the control group when the TNF does not bind TNFR. We used Edu technology to repeat the above experiments and the results were similar. Compared with the control group, the hemocytes stimulated by rTNF showed more significant proliferation, and the proliferation rate was significantly down-regulated after knocking down the EsTNFR gene. Therefore, we indicate that TNF binding TNFR can affect the proliferation of E. sinensis hemocytes, which might be manifested by affecting the expression of some proliferation-related genes.
Collapse
Affiliation(s)
- Yufan Bao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Guoqing Shen
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Yanan Guo
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China
| | - Xinpeng Fan
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China.
| | - Weiwei Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
37
|
Touré H, Herrmann JL, Szuplewski S, Girard-Misguich F. Drosophila melanogaster as an organism model for studying cystic fibrosis and its major associated microbial infections. Infect Immun 2023; 91:e0024023. [PMID: 37847031 PMCID: PMC10652941 DOI: 10.1128/iai.00240-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Cystic fibrosis (CF) is a human genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene that encodes a chloride channel. The most severe clinical manifestation is associated with chronic pulmonary infections by pathogenic and opportunistic microbes. Drosophila melanogaster has become the invertebrate model of choice for modeling microbial infections and studying the induced innate immune response. Here, we review its contribution to the understanding of infections with six major pathogens associated with CF (Staphylococcus aureus, Pseudomonas aeruginosa, Burkholderia cepacia, Mycobacterium abscessus, Streptococcus pneumoniae, and Aspergillus fumigatus) together with the perspectives opened by the recent availability of two CF models in this model organism.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| | - 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
| | - Sébastien Szuplewski
- Université Paris-Saclay, UVSQ, Laboratoire de Génétique et Biologie Cellulaire, Montigny-le-Bretonneux, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, Infection et Inflammation, Montigny-le-Bretonneux, France
| |
Collapse
|
38
|
Sun J, Zhang C, Gao F, Stathopoulos A. Single-cell transcriptomics illuminates regulatory steps driving anterior-posterior patterning of Drosophila embryonic mesoderm. Cell Rep 2023; 42:113289. [PMID: 37858470 DOI: 10.1016/j.celrep.2023.113289] [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: 03/28/2023] [Revised: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023] Open
Abstract
Single-cell technologies promise to uncover how transcriptional programs orchestrate complex processes during embryogenesis. Here, we apply a combination of single-cell technology and genetic analysis to investigate the dynamic transcriptional changes associated with Drosophila embryo morphogenesis at gastrulation. Our dataset encompassing the blastoderm-to-gastrula transition provides a comprehensive single-cell map of gene expression across cell lineages validated by genetic analysis. Subclustering and trajectory analyses revealed a surprising stepwise progression in patterning to transition zygotic gene expression and specify germ layers as well as uncovered an early role for ecdysone signaling in epithelial-to-mesenchymal transition in the mesoderm. We also show multipotent progenitors arise prior to gastrulation by analyzing the transcription trajectory of caudal mesoderm cells, including a derivative that ultimately incorporates into visceral muscles of the midgut and hindgut. This study provides a rich resource of gastrulation and elucidates spatially regulated temporal transitions of transcription states during the process.
Collapse
Affiliation(s)
- Jingjing Sun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Chen Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Fan Gao
- Bioinformatics Resource Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
39
|
Heron R, Amato C, Wood W, Davidson AJ. Understanding the diversity and dynamics of in vivo efferocytosis: Insights from the fly embryo. Immunol Rev 2023; 319:27-44. [PMID: 37589239 PMCID: PMC10952863 DOI: 10.1111/imr.13266] [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: 04/03/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
The clearance of dead and dying cells, termed efferocytosis, is a rapid and efficient process and one that is critical for organismal health. The extraordinary speed and efficiency with which dead cells are detected and engulfed by immune cells within tissues presents a challenge to researchers who wish to unravel this fascinating process, since these fleeting moments of uptake are almost impossible to catch in vivo. In recent years, the fruit fly (Drosophila melanogaster) embryo has emerged as a powerful model to circumvent this problem. With its abundance of dying cells, specialist phagocytes and relative ease of live imaging, the humble fly embryo provides a unique opportunity to catch and study the moment of cell engulfment in real-time within a living animal. In this review, we explore the recent advances that have come from studies in the fly, and how live imaging and genetics have revealed a previously unappreciated level of diversity in the efferocytic program. A variety of efferocytic strategies across the phagocytic cell population ensure efficient and rapid clearance of corpses wherever death is encountered within the varied and complex setting of a multicellular living organism.
Collapse
Affiliation(s)
- Rosalind Heron
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Clelia Amato
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Will Wood
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
| | - Andrew J. Davidson
- Institute for Regeneration and RepairUniversity of EdinburghEdinburghUK
- School of Cancer SciencesWolfson Wohl Cancer Research Centre, University of GlasgowGlasgowUK
| |
Collapse
|
40
|
Juarez-Carreño S, Geissmann F. The macrophage genetic cassette inr/dtor/pvf2 is a nutritional status checkpoint for developmental timing. SCIENCE ADVANCES 2023; 9:eadh0589. [PMID: 37729406 PMCID: PMC10511196 DOI: 10.1126/sciadv.adh0589] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/17/2023] [Indexed: 09/22/2023]
Abstract
A small number of signaling molecules, used reiteratively, control differentiation programs, but the mechanisms that adapt developmental timing to environmental cues are less understood. We report here that a macrophage inr/dtor/pvf2 genetic cassette is a developmental timing checkpoint in Drosophila, which either licenses or delays biosynthesis of the steroid hormone in the endocrine gland and metamorphosis according to the larval nutritional status. Insulin receptor/dTor signaling in macrophages is required and sufficient for production of the PDGF/VEGF family growth factor Pvf2, which turns on transcription of the sterol biosynthesis Halloween genes in the prothoracic gland via its receptor Pvr. In response to a starvation event or genetic manipulation, low Pvf2 signal delays steroid biosynthesis until it becomes Pvr-independent, thereby prolonging larval growth before pupariation. The significance of this developmental timing checkpoint for host fitness is illustrated by the observation that it regulates the size of the pupae and adult flies.
Collapse
|
41
|
Khalili D, Mohammed M, Kunc M, Sindlerova M, Ankarklev J, Theopold U. Single-cell sequencing of tumor-associated macrophages in a Drosophila model. Front Immunol 2023; 14:1243797. [PMID: 37795097 PMCID: PMC10546068 DOI: 10.3389/fimmu.2023.1243797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/31/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction Tumor-associated macrophages may act to either limit or promote tumor growth, yet the molecular basis for either path is poorly characterized. Methods We use a larval Drosophila model that expresses a dominant-active version of the Ras-oncogene (RasV12) to study dysplastic growth during early tumor progression. We performed single-cell RNA-sequencing of macrophage-like hemocytes to characterize these cells in tumor- compared to wild-type larvae. Hemocytes included manually extracted tumor-associated- and circulating cells. Results and discussion We identified five distinct hemocyte clusters. In addition to RasV12 larvae, we included a tumor model where the activation of effector caspases was inhibited, mimicking an apoptosis-resistant setting. Circulating hemocytes from both tumor models differ qualitatively from control wild-type cells-they display an enrichment for genes involved in cell division, which was confirmed using proliferation assays. Split analysis of the tumor models further reveals that proliferation is strongest in the caspase-deficient setting. Similarly, depending on the tumor model, hemocytes that attach to tumors activate different sets of immune effectors-antimicrobial peptides dominate the response against the tumor alone, while caspase inhibition induces a shift toward members of proteolytic cascades. Finally, we provide evidence for transcript transfer between hemocytes and possibly other tissues. Taken together, our data support the usefulness of Drosophila to study the response against tumors at the organismic level.
Collapse
Affiliation(s)
- Dilan Khalili
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| | - Mubasher Mohammed
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| | - Martin Kunc
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Martina Sindlerova
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| | - Johan Ankarklev
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| | - Ulrich Theopold
- The Wenner-Gren Institute, Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|
42
|
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
|
43
|
Rolandelli A, Laukaitis-Yousey HJ, Bogale HN, Singh N, Samaddar S, O’Neal AJ, Ferraz CR, Butnaru M, Mameli E, Xia B, Mendes MT, Butler LR, Marnin L, Cabrera Paz FE, Valencia LM, Rana VS, Skerry C, Pal U, Mohr SE, Perrimon N, Serre D, Pedra JH. Tick hemocytes have pleiotropic roles in microbial infection and arthropod fitness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555785. [PMID: 37693411 PMCID: PMC10491215 DOI: 10.1101/2023.08.31.555785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Uncovering the complexity of systems in non-model organisms is critical for understanding arthropod immunology. Prior efforts have mostly focused on Dipteran insects, which only account for a subset of existing arthropod species in nature. Here, we describe immune cells or hemocytes from the clinically relevant tick Ixodes scapularis using bulk and single cell RNA sequencing combined with depletion via clodronate liposomes, RNA interference, Clustered Regularly Interspaced Short Palindromic Repeats activation (CRISPRa) and RNA-fluorescence in situ hybridization (FISH). We observe molecular alterations in hemocytes upon tick infestation of mammals and infection with either the Lyme disease spirochete Borrelia burgdorferi or the rickettsial agent Anaplasma phagocytophilum. We predict distinct hemocyte lineages and reveal clusters exhibiting defined signatures for immunity, metabolism, and proliferation during hematophagy. Furthermore, we perform a mechanistic characterization of two I. scapularis hemocyte markers: hemocytin and astakine. Depletion of phagocytic hemocytes affects hemocytin and astakine levels, which impacts blood feeding and molting behavior of ticks. Hemocytin specifically affects the c-Jun N-terminal kinase (JNK) signaling pathway, whereas astakine alters hemocyte proliferation in I. scapularis. Altogether, we uncover the heterogeneity and pleiotropic roles of hemocytes in ticks and provide a valuable resource for comparative biology in arthropods.
Collapse
Affiliation(s)
- Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hanna J. Laukaitis-Yousey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Haikel N. Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anya J. O’Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Camila R. Ferraz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Baolong Xia
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - M. Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - L. Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Francy E. Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Luisa M. Valencia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vipin S. Rana
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Serre
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joao H.F. Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Mass E, Nimmerjahn F, Kierdorf K, Schlitzer A. Tissue-specific macrophages: how they develop and choreograph tissue biology. Nat Rev Immunol 2023; 23:563-579. [PMID: 36922638 PMCID: PMC10017071 DOI: 10.1038/s41577-023-00848-y] [Citation(s) in RCA: 232] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 03/17/2023]
Abstract
Macrophages are innate immune cells that form a 3D network in all our tissues, where they phagocytose dying cells and cell debris, immune complexes, bacteria and other waste products. Simultaneously, they produce growth factors and signalling molecules - such activities not only promote host protection in response to invading microorganisms but are also crucial for organ development and homeostasis. There is mounting evidence of macrophages orchestrating fundamental physiological processes, such as blood vessel formation, adipogenesis, metabolism and central and peripheral neuronal function. In parallel, novel methodologies have led to the characterization of tissue-specific macrophages, with distinct subpopulations of these cells showing different developmental trajectories, transcriptional programmes and life cycles. Here, we summarize our growing knowledge of macrophage diversity and how macrophage subsets orchestrate tissue development and function. We further interrelate macrophage ontogeny with their core functions across tissues, that is, the signalling events within the macrophage niche that may control organ functionality during development, homeostasis and ageing. Finally, we highlight the open questions that will need to be addressed by future studies to better understand the tissue-specific functions of distinct macrophage subsets.
Collapse
Affiliation(s)
- Elvira Mass
- Developmental Biology of the Immune System, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Centre for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
45
|
Hopkins BR, Barmina O, Kopp A. A single-cell atlas of the sexually dimorphic Drosophila foreleg and its sensory organs during development. PLoS Biol 2023; 21:e3002148. [PMID: 37379332 DOI: 10.1371/journal.pbio.3002148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/03/2023] [Indexed: 06/30/2023] Open
Abstract
To respond to the world around them, animals rely on the input of a network of sensory organs distributed throughout the body. Distinct classes of sensory organs are specialized for the detection of specific stimuli such as strain, pressure, or taste. The features that underlie this specialization relate both to the neurons that innervate sensory organs and the accessory cells they comprise. To understand the genetic basis of this diversity of cell types, both within and between sensory organs, we performed single-cell RNA sequencing on the first tarsal segment of the male Drosophila melanogaster foreleg during pupal development. This tissue displays a wide variety of functionally and structurally distinct sensory organs, including campaniform sensilla, mechanosensory bristles, and chemosensory taste bristles, as well as the sex comb, a recently evolved male-specific structure. In this study, we characterize the cellular landscape in which the sensory organs reside, identify a novel cell type that contributes to the construction of the neural lamella, and resolve the transcriptomic differences among support cells within and between sensory organs. We identify the genes that distinguish between mechanosensory and chemosensory neurons, resolve a combinatorial transcription factor code that defines 4 distinct classes of gustatory neurons and several types of mechanosensory neurons, and match the expression of sensory receptor genes to specific neuron classes. Collectively, our work identifies core genetic features of a variety of sensory organs and provides a rich, annotated resource for studying their development and function.
Collapse
Affiliation(s)
- Ben R Hopkins
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| | - Olga Barmina
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California, Davis, California, United States of America
| |
Collapse
|
46
|
Wrońska AK, Kaczmarek A, Boguś MI, Kuna A. Lipids as a key element of insect defense systems. Front Genet 2023; 14:1183659. [PMID: 37359377 PMCID: PMC10289264 DOI: 10.3389/fgene.2023.1183659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
The relationship between insect pathogenic fungi and their insect hosts is a classic example of a co-evolutionary arms race between pathogen and target host: parasites evolve towards mechanisms that increase their advantage over the host, and the host increasingly strengthens its defenses. The present review summarizes the literature data describing the direct and indirect role of lipids as an important defense mechanism during fungal infection. Insect defense mechanisms comprise anatomical and physiological barriers, and cellular and humoral response mechanisms. The entomopathogenic fungi have the unique ability to digest the insect cuticle by producing hydrolytic enzymes with chitin-, lipo- and proteolytic activity; besides the oral tract, cuticle pays the way for fungal entry within the host. The key factor in insect resistance to fungal infection is the presence of certain types of lipids (free fatty acids, waxes or hydrocarbons) which can promote or inhibit fungal attachment to cuticle, and might also have antifungal activity. Lipids are considered as an important source of energy, and as triglycerides are stored in the fat body, a structure analogous to the liver and adipose tissue in vertebrates. In addition, the fat body plays a key role in innate humoral immunity by producing a range of bactericidal proteins and polypeptides, one of which is lysozyme. Energy derived from lipid metabolism is used by hemocytes to migrate to the site of fungal infection, and for phagocytosis, nodulation and encapsulation. One polyunsaturated fatty acid, arachidonic acid, is used in the synthesis of eicosanoids, which play several crucial roles in insect physiology and immunology. Apolipoprotein III is important compound with antifungal activity, which can modulate insect cellular response and is considered as important signal molecule.
Collapse
Affiliation(s)
- Anna Katarzyna Wrońska
- Museum and Institute of Zoology, Polish Academy of Science, Warszawa, Poland
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Kaczmarek
- Museum and Institute of Zoology, Polish Academy of Science, Warszawa, Poland
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Mieczysława Irena Boguś
- Museum and Institute of Zoology, Polish Academy of Science, Warszawa, Poland
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Kuna
- Independent Researcher, Warsaw, Poland
| |
Collapse
|
47
|
Chung HL, Ye Q, Park YJ, Zuo Z, Mok JW, Kanca O, Tattikota SG, Lu S, Perrimon N, Lee HK, Bellen HJ. Very-long-chain fatty acids induce glial-derived sphingosine-1-phosphate synthesis, secretion, and neuroinflammation. Cell Metab 2023; 35:855-874.e5. [PMID: 37084732 PMCID: PMC10160010 DOI: 10.1016/j.cmet.2023.03.022] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
VLCFAs (very-long-chain fatty acids) are the most abundant fatty acids in myelin. Hence, during demyelination or aging, glia are exposed to higher levels of VLCFA than normal. We report that glia convert these VLCFA into sphingosine-1-phosphate (S1P) via a glial-specific S1P pathway. Excess S1P causes neuroinflammation, NF-κB activation, and macrophage infiltration into the CNS. Suppressing the function of S1P in fly glia or neurons, or administration of Fingolimod, an S1P receptor antagonist, strongly attenuates the phenotypes caused by excess VLCFAs. In contrast, elevating the VLCFA levels in glia and immune cells exacerbates these phenotypes. Elevated VLCFA and S1P are also toxic in vertebrates based on a mouse model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). Indeed, reducing VLCFA with bezafibrate ameliorates the phenotypes. Moreover, simultaneous use of bezafibrate and fingolimod synergizes to improve EAE, suggesting that lowering VLCFA and S1P is a treatment avenue for MS.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Qi Ye
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nobert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hyun Kyoung Lee
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
48
|
Tian Y, Morin-Poulard I, Liu X, Vanzo N, Crozatier M. A mechanosensitive vascular niche for Drosophila hematopoiesis. Proc Natl Acad Sci U S A 2023; 120:e2217862120. [PMID: 37094122 PMCID: PMC10160988 DOI: 10.1073/pnas.2217862120] [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: 10/19/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
Hematopoietic stem and progenitor cells maintain blood cell homeostasis by integrating various cues provided by specialized microenvironments or niches. Biomechanical forces are emerging as key regulators of hematopoiesis. Here, we report that mechanical stimuli provided by blood flow in the vascular niche control Drosophila hematopoiesis. In vascular niche cells, the mechanosensitive channel Piezo transduces mechanical forces through intracellular calcium upregulation, leading to Notch activation and repression of FGF ligand transcription, known to regulate hematopoietic progenitor maintenance. Our results provide insight into how the vascular niche integrates mechanical stimuli to regulate hematopoiesis.
Collapse
Affiliation(s)
- Yushun Tian
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Ismaël Morin-Poulard
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Xiaohui Liu
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Nathalie Vanzo
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| | - Michèle Crozatier
- Molecular, Cellular, and Development/UMR5077, Centre de Biologie Intégrative, Toulouse Cedex 931062, France
| |
Collapse
|
49
|
Bland ML. Regulating metabolism to shape immune function: Lessons from Drosophila. Semin Cell Dev Biol 2023; 138:128-141. [PMID: 35440411 PMCID: PMC10617008 DOI: 10.1016/j.semcdb.2022.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/21/2022] [Accepted: 04/03/2022] [Indexed: 12/14/2022]
Abstract
Infection with pathogenic microbes is a severe threat that hosts manage by activating the innate immune response. In Drosophila melanogaster, the Toll and Imd signaling pathways are activated by pathogen-associated molecular patterns to initiate cellular and humoral immune processes that neutralize and kill invaders. The Toll and Imd signaling pathways operate in organs such as fat body and gut that control host nutrient metabolism, and infections or genetic activation of Toll and Imd signaling also induce wide-ranging changes in host lipid, carbohydrate and protein metabolism. Metabolic regulation by immune signaling can confer resistance to or tolerance of infection, but it can also lead to pathology and susceptibility to infection. These immunometabolic phenotypes are described in this review, as are changes in endocrine signaling and gene regulation that mediate survival during infection. Future work in the field is anticipated to determine key variables such as sex, dietary nutrients, life stage, and pathogen characteristics that modify immunometabolic phenotypes and, importantly, to uncover the mechanisms used by the immune system to regulate metabolism.
Collapse
Affiliation(s)
- Michelle L Bland
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, United States.
| |
Collapse
|
50
|
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: 14] [Impact Index Per Article: 7.0] [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
|