1
|
Ghosh G, Das D, Nandi A, De S, Gangappa SN, Prasad M. Ecdysone regulates phagocytic cell fate of epithelial cells in developing Drosophila eggs. J Cell Biol 2025; 224:e202411073. [PMID: 40434296 PMCID: PMC12118371 DOI: 10.1083/jcb.202411073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/01/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Acquisition of nonprofessional phagocytic cell fate plays an important role in sculpting functional metazoan organs and maintaining overall tissue homeostasis. Though physiologically highly relevant, how the normal epithelial cells acquire phagocytic fate is still mostly unclear. We have employed the Drosophila ovary model to demonstrate that the classical ecdysone signaling in the somatic epithelial follicle cells (AFCs) aids the removal of germline nurse cells (NCs) in late oogenesis. Our live-cell imaging data reveal a novel phenomenon wherein collective behavior of 4-5 AFCs is required for clearing a single NC. By employing classical genetics, molecular biology, and yeast one-hybrid assay, we demonstrate that ecdysone modulates the phagocytic disposition of AFCs at two levels. It regulates the epithelial-mesenchymal transition of the AFCs through Serpent and modulates the phagocytic behavior of the AFCs through Croquemort and Draper. Our data provide unprecedented novel molecular insights into how ecdysone signaling reprograms AFCs toward a phagocytic fate.
Collapse
Affiliation(s)
- Gaurab Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Devyan Das
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Abhrajyoti Nandi
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Souvik De
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Sreeramaiah N. Gangappa
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| | - Mohit Prasad
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, India
| |
Collapse
|
2
|
Touré H, Durand N, Orgeur M, Galindo LA, Girard-Misguich F, Guénal I, Herrmann JL, Szuplewski S. ENaC is a host susceptibility factor to bacterial infections in cystic fibrosis context. Commun Biol 2025; 8:653. [PMID: 40269088 PMCID: PMC12019357 DOI: 10.1038/s42003-025-07877-4] [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: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 04/25/2025] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by dysfunction in the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) chloride channel. Patients with CF are hypersusceptible to Mycobacterium abscessus infection, a fast-growing mycobacterium and harmful opportunistic pathogen. Although CFTR dysfunction is known as a host susceptibility factor for M. abscessus infection, the functional impact of the trimeric Epithelial sodium Channel (ENaC), whose activity is negatively regulated by CFTR, towards M. abscessus infection has not been explored yet. To address this issue, we took advantage of miR-263a deficient Drosophila presenting a CF-like phenotype due to ENaC hyperactivity (ENaC+ ). We observed that the ENaC+ flies were as hypersusceptible to M. abscessus infection as the Cftr-deficient flies. The hypersensitivity of ENaC+ flies to M. abscessus infection was fully rescued by blocking ENaC hyperactivity, both chemically and genetically. Furthermore, we observed that ENaC hyperactivity per se was detrimental to ENaC+ Drosophila, as they were unable to mount an efficient humoral immune response. Upon infection, ENaC+ flies failed to upregulate 20-hydroxyecdysone production, which subsequently altered the production of protective antimicrobial peptides against M. abscessus. Overall, our results show that ENaC plays a key role in host susceptibility to M. abscessus infection and, correlatively to other CF pathogens.
Collapse
Affiliation(s)
- Hamadoun Touré
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Nicolas Durand
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France
| | - Mickael Orgeur
- Institut Pasteur, Université Paris Cité, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Lee Ann Galindo
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France
- The American University of Paris, Paris, France
| | - Fabienne Girard-Misguich
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France
| | - Isabelle Guénal
- Université Paris-Saclay, UVSQ, EA 4589 Laboratoire de Génétique et Biologie Cellulaire, Montigny-le-Bretonneux, France
| | - Jean-Louis Herrmann
- Université Paris-Saclay, UVSQ, INSERM, U1173 Infection et Inflammation, Montigny-le-Bretonneux, France.
- Assistance Publique - Hôpitaux de Paris, GHU Paris-Saclay, Hôpital Raymond Poincaré, Garches, France.
| | - Sébastien Szuplewski
- Université Paris-Saclay, UVSQ, EA 4589 Laboratoire de Génétique et Biologie Cellulaire, Montigny-le-Bretonneux, France.
| |
Collapse
|
3
|
Leyria J, Fruttero LL, Paglione PA, Canavoso LE. How Insects Balance Reproductive Output and Immune Investment. INSECTS 2025; 16:311. [PMID: 40266843 PMCID: PMC11943238 DOI: 10.3390/insects16030311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 04/25/2025]
Abstract
Insects face the constant challenge of balancing energy allocation between reproduction and immune responses, both of which are highly energy-demanding processes. Immune challenges frequently result in decreased fecundity, reduced egg viability, and delayed ovarian development. Conversely, heightened reproductive activity often suppresses immune functions. This trade-off has profound ecological and evolutionary consequences, shaping insects' survival, adaptation, and population dynamics. The intricate interplay between reproduction and immunity in insects is regulated by the neuroendocrine and endocrine systems, which orchestrate resource distribution alongside other biological processes. Key hormones, such as juvenile hormone and ecdysteroids, serve as central regulators, influencing both immune responses and reproductive activities. Additionally, macromolecules like vitellogenin and lipophorin, primarily known for their functions as yolk protein precursors and lipid carriers, play crucial roles in pathogen recognition and transgenerational immune priming. Advancements in molecular and omics tools have unveiled the complexity of these regulatory mechanisms, providing new insights into how insects dynamically allocate resources to optimize their fitness. This delicate balance underscores critical evolutionary strategies and the integration of physiological systems across species. This review synthesizes insights from life history theory, oogenesis, and immunity, offering new perspectives on the trade-offs between reproductive output and immune investment.
Collapse
Affiliation(s)
- Jimena Leyria
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, CP, Argentina; (J.L.); (L.L.F.); (P.A.P.)
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, CP, Argentina
| | - Leonardo L. Fruttero
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, CP, Argentina; (J.L.); (L.L.F.); (P.A.P.)
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, CP, Argentina
| | - Pedro A. Paglione
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, CP, Argentina; (J.L.); (L.L.F.); (P.A.P.)
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, CP, Argentina
| | - Lilián E. Canavoso
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba 5000, CP, Argentina; (J.L.); (L.L.F.); (P.A.P.)
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, CP, Argentina
| |
Collapse
|
4
|
Xu P, He Z, Gao X, Zeng X, Wei D, Long X, Yu Y. Research on the Expression of Immune-Related Genes at Different Stages in the Third-Instar Larvae of Spodoptera frugiperda Infected by Metarhizium rileyi. INSECTS 2025; 16:199. [PMID: 40003829 PMCID: PMC11856804 DOI: 10.3390/insects16020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
Spodoptera frugiperda is a major migratory agricultural pest that poses a significant threat to global crop safety. Metarhizium rileyi has emerged as an effective biocontrol agent against lepidopteran pests. In this study, we examined the immune responses of third-instar S. frugiperda larvae at various stages of an M. rileyi infection. Using RNA-seq and microscopic observation, we identified the immune-related pathways enriched at different infection stages, which were further validated by a qRT-PCR. Our findings revealed the following immune responses during infection: During the stage when M. rileyi penetrated the host cuticle (0-48 h), the genes related to energy metabolism, detoxification, and melanization were upregulated. Meanwhile, the TOLL and IMD signaling pathways were activated to counter the infection. During the stage of M. rileyi's internal infection (48-96 h), which was the peak expression period of the immune-related genes, cellular immunity predominated. Hemocytes encapsulated and phagocytosed the hyphal bodies. Phagocytosis was enhanced through the upregulation of the genes related to ROS and the melanization-related genes, as well as the genes involved in insect hormone biosynthesis. During the stage when M. rileyi grew from the inside to the outside of the host (96-120 h), immune system paralysis resulted in host mortality. These findings deepen our understanding of the immune interactions between M. rileyi and S. frugiperda, support the potential of M. rileyi as an effective biocontrol agent, and provide a theoretical foundation for the development of targeted biopesticides for pests using biotechnological approaches.
Collapse
Affiliation(s)
- Pengfei Xu
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Zhan He
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Xuyuan Gao
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Xianru Zeng
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Dewei Wei
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Xiuzhen Long
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| | - Yonghao Yu
- Key Laboratory of Green Prevention and Control on Fruits and Vegetables in South China Ministry of Agriculture and Rural Affairs, Nanning 530007, China; (P.X.); (Z.H.); (X.G.); (X.Z.); (D.W.)
- Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China
- Plant Protection Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China
| |
Collapse
|
5
|
Vieira CS, Bisogno S, Salvemini M, Loza Telleria E, Volf P. Azadirachtin disrupts ecdysone signaling and alters sand fly immunity. Parasit Vectors 2024; 17:526. [PMID: 39707409 DOI: 10.1186/s13071-024-06589-8] [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: 09/03/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Leishmaniasis is a group of neglected vector-borne diseases transmitted by phlebotomine sand flies. Leishmania parasites must overcome various defenses in the sand fly midgut, including the insects's immune response. Insect immunity is regulated by the ecdysone hormone, which binds to its nuclear receptor (EcR) and activates the transcription of genes involved in insect immunity. However, the role of ecdysone in sand fly immunity has never been studied. Phlebotomus perniciosus is a natural vector of Leishmania infantum; here, we manipulated its neuroendocrine system using azadirachtin (Aza), a natural compound known to affect ecdysone synthesis. METHODS Phlebotomus perniciosus larvae and adult females were fed on food containing either Aza alone or Aza plus ecdysone, and the effects on mortality and ecdysis were evaluated. Genes related to ecdysone signaling and immunity were identified in P. perniciosus, and the expression of antimicrobial peptides (AMPs), EcR, the ecdysone-induced genes Eip74EF and Eip75B, and the transcription factor serpent were analyzed using quantitative polymerase chain reaction (PCR). RESULTS Aza treatment inhibited molting of first-instar (L1) larvae to L2, with only 10% of larvae molting compared to 95% in the control group. Serpent and Eip74EF, attacin, defensin 1, and defensin 2 genes were downregulated by Aza treatment in larvae. Similarly, Aza-treated adult females also presented suppression of ecdysone signaling-related genes and the AMPs attacin and defensin 2. Notably, all gene repression caused by Aza was reversed by adding ecdysone concomitantly with Aza to the larval or female food, indicating that these genes are effective markers for ecdysone repression. CONCLUSIONS These results highlight the critical role of ecdysone in regulating the development and immunity of P. perniciosus, which potentially could interfere with Leishmania infection.
Collapse
Affiliation(s)
- Cecilia Stahl Vieira
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Sara Bisogno
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marco Salvemini
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Erich Loza Telleria
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
6
|
Kwon SY, Chan K, Stofanko M, Chan KH, Badenhorst P. Abrupt-mediated control of ninjurins regulates Drosophila sessile haemocyte compartments. Development 2024; 151:dev202977. [PMID: 39545919 DOI: 10.1242/dev.202977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Macrophage-like cells called haemocytes are key effectors of Drosophila cellular innate immune function. Larval haemocytes exist either in circulation or localize to segmentally repeated sessile haemocyte compartments (SHCs). While numerous functions have been proposed for SHCs, the mechanisms directing haemocytes to them are unclear. Here, we have exploited the developmentally regulated dispersal of SHCs that occurs at pupariation to identify the Abrupt (Ab) transcription factor (TF) and ninjurin cell-adhesion molecules as regulators of haemocyte recruitment to SHCs. We show that larval haemocytes express ninjurins, which are required for targeting haemocytes to SHCs. However, at pupariation, ecdysteroid signalling stimulates Ab expression, which collaborates with TFs, including Blimp-1 and Hr3, to repress ninjurins and disperse haemocytes. We observe that experimental manipulations that antagonize ninjurin function in larval haemocytes cause premature SHC dispersal, while stabilization of ninjurins in haemocytes blocks developmentally regulated SHC remodelling and increases sensitivity to immune challenges. Cumulatively, our data indicate that control of ninjurin activity provides a common target through which diverse developmental, environmental and immune stimuli can be integrated to control haemocyte dispersal and immune function.
Collapse
Affiliation(s)
- So Yeon Kwon
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Kimberly Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Martin Stofanko
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Ka Hei Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Paul Badenhorst
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
7
|
Vidal M, Arch M, Fuentes E, Cardona PJ. Drosophila melanogaster experimental model to test new antimicrobials: a methodological approach. Front Microbiol 2024; 15:1478263. [PMID: 39568995 PMCID: PMC11576456 DOI: 10.3389/fmicb.2024.1478263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
Given the increasing concern about antimicrobial resistance among the microorganisms that cause infections in our society, there is an urgent need for new drug discovery. Currently, this process involves testing many low-quality compounds, resulting from the in vivo testing, on mammal models, which not only wastes time, resources, and money, but also raises ethical questions. In this review, we have discussed the potential of D. melanogaster as an intermediary experimental model in this drug discovery timeline. We have tackled the topic from a methodological perspective, providing recommendations regarding the range of drug concentrations to test based on the mechanism of action of each compound; how to treat D. melanogaster, how to monitor that treatment, and what parameters we should consider when designing a drug screening protocol to maximize the study's benefits. We also discuss the necessary improvements needed to establish the D. melanogaster model of infection as a standard technique in the drug screening process. Overall, D. melanogaster has been demonstrated to be a manageable model for studying broad-spectrum infection treatment. It allows us to obtain valuable information in a cost-effective manner, which can improve the drug screening process and provide insights into our current major concern. This approach is also in line with the 3R policy in biomedical research, in particular on the replacement and reduce the use of vertebrates in preclinical development.
Collapse
Affiliation(s)
- Maria Vidal
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Marta Arch
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Esther Fuentes
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
| | - Pere-Joan Cardona
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital (HUGTP), Badalona, Catalonia, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
8
|
Schweibenz CK, Placentra VC, Moberg KH. The Drosophila EcR-Hippo component Taiman promotes epithelial cell fitness by control of the Dally-like glypican and Wg gradient. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.31.587486. [PMID: 38617327 PMCID: PMC11014482 DOI: 10.1101/2024.03.31.587486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Rapidly dividing cells can eliminate slow growing neighbors through the apoptotic process of cell competition. This process ensures that only high fitness cells populate embryonic tissues and is proposed to underlie the ability of oncogene-transformed cells to progressively replace normal cells within a tissue. Patches of cells in the Drosophila wing disc overexpressing the oncogenic Taiman (Tai) transcriptional coactivator kill normal neighbors by secreting Spz ligands that trigger pro-apoptotic Toll signaling in receiving cells. However, extracellular signaling mechanisms responsible for elimination of slow growing cells by normal neighbors remain poorly defined. Here we show that slow growing cells with reduced Tai (Tailow) are killed by normal neighbors through a mechanism involving competition for the Wingless (Wg/Wnt) ligand. Elevated Wg signaling significantly rescues elimination of Tailow cells in multiple organs, suggesting that Tai may normally promote Wg activity. Examining distribution of Wg components reveals that Tai promotes extracellular spread of the Wg ligand from source cells across the wing disc, thus ensuring patterned expression of multiple Wg-regulated target genes. Tai controls Wg spread indirectly through the extracellular glypican Dally-like protein (Dlp), which binds Wg and promotes its extracellular diffusion and capture by receptors. Data indicate that Tai likely controls Dlp at two levels: transcription of dlp mRNA and Dlp intracellular trafficking. Overall, these data indicate that the Tai acts through Dlp to enable Wg transport and signaling, and that cell competition in the Tailow model arises due to inequity in the ability of epithelial cells to sequester limiting amounts of the Wg growth factor.
Collapse
Affiliation(s)
- Colby K. Schweibenz
- Department of Cell Biology, Emory University School of Medicine
- Graduate Program in Biochemistry, Cell, and Developmental Biology
| | - Victoria C. Placentra
- Department of Cell Biology, Emory University School of Medicine
- Graduate Program in Genetics and Molecular Biology, Emory University
| | | |
Collapse
|
9
|
Lansdon P, Kasuya J, Kitamoto T. Commensal bacteria exacerbate seizure-like phenotypes in Drosophila voltage-gated sodium channel mutants. GENES, BRAIN, AND BEHAVIOR 2024; 23:e70000. [PMID: 39231190 PMCID: PMC11373613 DOI: 10.1111/gbb.70000] [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: 03/15/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
Mutations in voltage-gated sodium (Nav) channels, which are essential for generating and propagating action potentials, can lead to serious neurological disorders, such as epilepsy. However, disease-causing Nav channel mutations do not always result in severe symptoms, suggesting that the disease conditions are significantly affected by other genetic factors and various environmental exposures, collectively known as the "exposome". Notably, recent research emphasizes the pivotal role of commensal bacteria in neural development and function. Although these bacteria typically benefit the nervous system under normal conditions, their impact during pathological states remains largely unknown. Here, we investigated the influence of commensal microbes on seizure-like phenotypes exhibited by paraShu-a gain-of-function mutant of the Drosophila Nav channel gene, paralytic. Remarkably, the elimination of endogenous bacteria considerably ameliorated neurological impairments in paraShu. Consistently, reintroducing bacteria, specifically from the Lactobacillus or Acetobacter genera, heightened the phenotypic severity in the bacteria-deprived mutants. These findings posit that particular native bacteria contribute to the severity of seizure-like phenotypes in paraShu. We further uncovered that treating paraShu with antibiotics boosted Nrf2 signaling in the gut, and that global Nrf2 activation mirrored the effects of removing bacteria from paraShu. This raises the possibility that the removal of commensal bacteria suppresses the seizure-like manifestations through augmented antioxidant responses. Since bacterial removal during development was critical for suppression of adult paraShu phenotypes, our research sets the stage for subsequent studies, aiming to elucidate the interplay between commensal bacteria and the developing nervous system in conditions predisposed to the hyperexcitable nervous system.
Collapse
Affiliation(s)
- Patrick Lansdon
- Interdisciplinary Graduate Program in Genetics, Graduate College, University of Iowa, Iowa City, Iowa, USA
| | - Junko Kasuya
- Department of Anesthesia, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Toshihiro Kitamoto
- Interdisciplinary Graduate Program in Genetics, Graduate College, University of Iowa, Iowa City, Iowa, USA
- Department of Anesthesia, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
10
|
Li J, Ma Y, Wu Z, Li J, Wang F, Yang Z, Xi Y, Yang D, Jiang Y, Yi Q, Huang S. The involvement of tumor necrosis factor receptor-associated factor 6 in regulating immune response by NF-κB at pre-molt stage of Chinese mitten crab (Eriocheir sinensis). FISH & SHELLFISH IMMUNOLOGY 2024; 153:109842. [PMID: 39153580 DOI: 10.1016/j.fsi.2024.109842] [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: 05/28/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Molting is a crucial biological process of crustaceans. Crustaceans go through three separate stages throughout their molting process, including pre-molt, post-molt and inter-molt. However, the exact mechanism of immunological modulation during molting remains unclear. Tumor necrosis factor receptor-associated factor 6 (TRAF6) has been extensively documented to participate in immune defense. In the present study, a TRAF6 gene with two TRAF-type zinc finger domains was identified from Eriocheir sinensis (designed as EsTRAF6), and its role in regulating immune response during molting process was explored. The mRNA expression level of EsTRAF6 at pre-molt stage was higher than that at post-molt stage and inter-molt stage. After Aeromonas hydrophila stimulation, the expression levels of EsTRAF6, EsRelish and anti-lipopolysaccharide factors (ALFs) genes exhibited a considerable increase at three molting stages. Subsequently, the expression patterns of EsTRAF6 and EsRelish in response to the treatment with 20-hydroxyecdysone (20E) were examined. The mRNA expression of EsTRAF6 and EsRelish were significantly increased at 12 h after 20E injection. Additionally, the protein expression level of TRAF6 was also up-regulated in 20E group compared to control group. Furthermore, the role of EsTRAF6 in regulating the anti- ALFs expression at pre-molt stage post A. hydrophila stimulation was investigated. Following the inhibition of the EsTRAF6 transcript using RNAi or the injection of inhibitor (TMBPS), there was a notable decrease of the EsALF1, EsALF2 and EsALF3 transcripts. Moreover, a significant reduction in the phosphorylation level of NF-κB at pre-molt stage was observed after A. hydrophila stimulation in TRAF6-inhibited crabs. Collectively, our results suggest that EsTRAF6 could be induced by 20E and promoted the EsALFs expression by activating NF-κB at pre-molt stage, which provides a novel insight into the research of immune regulatory mechanism during the process of molting of crustaceans.
Collapse
Affiliation(s)
- Jialin Li
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Yuhan Ma
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Zihao Wu
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Jiaming Li
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Fengchi Wang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Zhichao Yang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Yuting Xi
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Dazuo Yang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Key Laboratory of Marine Bio-Resources Restoration and Habitat Reparation in Liaoning Province, Dalian Ocean University, Dalian, 116023, China
| | - Yusheng Jiang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Dalian Key Laboratory of Breeding, Reproduction and Aquaculture of Crustaceans, Dalian, 116023, China
| | - Qilin Yi
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China.
| | - Shu Huang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Key Laboratory of Marine Bio-Resources Restoration and Habitat Reparation in Liaoning Province, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Breeding, Reproduction and Aquaculture of Crustaceans, Dalian, 116023, China.
| |
Collapse
|
11
|
Galambos N, Vincent-Monegat C, Vallier A, Parisot N, Heddi A, Zaidman-Rémy A. Cereal weevils' antimicrobial peptides: at the crosstalk between development, endosymbiosis and immune response. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230062. [PMID: 38497254 PMCID: PMC10945404 DOI: 10.1098/rstb.2023.0062] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/12/2023] [Indexed: 03/19/2024] Open
Abstract
Interactions between animals and microbes are ubiquitous in nature and strongly impact animal physiology. These interactions are shaped by the host immune system, which responds to infections and contributes to tailor the associations with beneficial microorganisms. In many insects, beneficial symbiotic associations not only include gut commensals, but also intracellular bacteria, or endosymbionts. Endosymbionts are housed within specialized host cells, the bacteriocytes, and are transmitted vertically across host generations. Host-endosymbiont co-evolution shapes the endosymbiont genome and host immune system, which not only fights against microbial intruders, but also ensures the preservation of endosymbionts and the control of their load and location. The cereal weevil Sitophilus spp. is a remarkable model in which to study the evolutionary adaptation of the immune system to endosymbiosis owing to its binary association with a unique, relatively recently acquired nutritional endosymbiont, Sodalis pierantonius. This Gram-negative bacterium has not experienced the genome size shrinkage observed in long-term endosymbioses and has retained immunogenicity. We focus here on the sixteen antimicrobial peptides (AMPs) identified in the Sitophilus oryzae genome and their expression patterns in different tissues, along host development or upon immune challenges, to address their potential functions in the defensive response and endosymbiosis homeostasis along the insect life cycle. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- N. Galambos
- INSA Lyon, INRAE, BF2I, UMR203, 69621 Villeurbanne, France
| | | | - A. Vallier
- INRAE, INSA Lyon, BF2I, UMR203, 69621 Villeurbanne, France
| | - N. Parisot
- INSA Lyon, INRAE, BF2I, UMR203, 69621 Villeurbanne, France
| | - A. Heddi
- INSA Lyon, INRAE, BF2I, UMR203, 69621 Villeurbanne, France
| | - A. Zaidman-Rémy
- INSA Lyon, INRAE, BF2I, UMR203, 69621 Villeurbanne, France
- Institut universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
12
|
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
|
13
|
Zhou L, Meng G, Zhu L, Ma L, Chen K. Insect Antimicrobial Peptides as Guardians of Immunity and Beyond: A Review. Int J Mol Sci 2024; 25:3835. [PMID: 38612644 PMCID: PMC11011964 DOI: 10.3390/ijms25073835] [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: 01/19/2024] [Revised: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Antimicrobial peptides (AMPs), as immune effectors synthesized by a variety of organisms, not only constitute a robust defense mechanism against a broad spectrum of pathogens in the host but also show promising applications as effective antimicrobial agents. Notably, insects are significant reservoirs of natural AMPs. However, the complex array of variations in types, quantities, antimicrobial activities, and production pathways of AMPs, as well as evolution of AMPs across insect species, presents a significant challenge for immunity system understanding and AMP applications. This review covers insect AMP discoveries, classification, common properties, and mechanisms of action. Additionally, the types, quantities, and activities of immune-related AMPs in each model insect are also summarized. We conducted the first comprehensive investigation into the diversity, distribution, and evolution of 20 types of AMPs in model insects, employing phylogenetic analysis to describe their evolutionary relationships and shed light on conserved and distinctive AMP families. Furthermore, we summarize the regulatory pathways of AMP production through classical signaling pathways and additional pathways associated with Nitric Oxide, insulin-like signaling, and hormones. This review advances our understanding of AMPs as guardians in insect immunity systems and unlocks a gateway to insect AMP resources, facilitating the use of AMPs to address food safety concerns.
Collapse
Affiliation(s)
- Lizhen Zhou
- Department of Plant Protection, College of Plant Protection, Yangzhou University, Yangzhou 225009, China;
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China
| | - Guanliang Meng
- Zoological Research Museum Alexander Koenig, Leibniz Institute for the Analysis of Biodiversity Change, 53113 Bonn, Germany;
| | - Ling Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Li Ma
- College of Plant Protection, Shanxi Agricultural University, Taigu 030810, China
| | - Kangkang Chen
- Department of Plant Protection, College of Plant Protection, Yangzhou University, Yangzhou 225009, China;
| |
Collapse
|
14
|
Li B, Wang D, Xie X, Chen X, Liang G, Xing D, Zhao T, Wu J, Zhou X, Li C. Mosquito E-20-Monooxygenase Gene Knockout Increases Dengue Virus Replication in Aedes aegypti Cells. Viruses 2024; 16:525. [PMID: 38675868 PMCID: PMC11054288 DOI: 10.3390/v16040525] [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: 02/28/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
E-20-monooxygenase (E20MO) is an enzymatic product of the shade (shd) locus (cytochrome p450, E20MO). Initially discovered in Drosophila, E20MO facilitates the conversion of ecdysone (E) into 20-hydroxyecdysone (20E) and is crucial for oogenesis. Prior research has implicated 20E in growth, development, and insecticide resistance. However, little attention has been given to the association between the E20MO gene and DENV2 infection. The transcriptome of Ae. aegypti cells (Aag2 cells) infected with DENV2 revealed the presence of the E20MO gene. The subsequent quantification of E20MO gene expression levels in Aag2 cells post-DENV infection was carried out. A CRISPR/Cas9 system was utilized to create an E20MO gene knockout cell line (KO), which was then subjected to DENV infection. Analyses of DENV2 copies in KO and wild-type (WT) cells were conducted at different days post-infection (dpi). Plasmids containing E20MO were constructed and transfected into KO cells, with pre- and post-transfection viral copy comparisons. Gene expression levels of E20MO increased after DENV infection. Subsequently, a successful generation of an E20MO gene knockout cell line and the verification of code-shifting mutations at both DNA and RNA levels were achieved. Furthermore, significantly elevated DENV2 RNA copies were observed in the mid-infection phase for the KO cell line. Viral RNA copies were lower in cells transfected with plasmids containing E20MO, compared to KO cells. Through knockout and plasmid complementation experiments in Aag2 cells, the role of E20MO in controlling DENV2 replication was demonstrated. These findings contribute to our understanding of the intricate biological interactions between mosquitoes and arboviruses.
Collapse
Affiliation(s)
- Bo Li
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Di Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaoxue Xie
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiaoli Chen
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Guorui Liang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Dan Xing
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Teng Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jiahong Wu
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Xinyu Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Chunxiao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
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
|
18
|
Arch M, Vidal M, Fuentes E, Abat AS, Cardona PJ. The reproductive status determines tolerance and resistance to Mycobacterium marinum in Drosophila melanogaster. Evol Med Public Health 2023; 11:332-347. [PMID: 37868078 PMCID: PMC10590161 DOI: 10.1093/emph/eoad029] [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: 01/14/2023] [Revised: 07/27/2023] [Indexed: 10/24/2023] Open
Abstract
Sex and reproductive status of the host have a major impact on the immune response against infection. Our aim was to understand their impact on host tolerance or resistance in the systemic Mycobacterium marinum infection of Drosophila melanogaster. We measured host survival and bacillary load at time of death, as well as expression by quantitative real-time polymerase chain reaction of immune genes (diptericin and drosomycin). We also assessed the impact of metabolic and hormonal regulation in the protection against infection by measuring expression of upd3, impl2 and ecR. Our data showed increased resistance in actively mating flies and in mated females, while reducing their tolerance to infection. Data suggests that Toll and immune deficiency (Imd) pathways determine tolerance and resistance, respectively, while higher basal levels of ecR favours the stimulation of the Imd pathway. A dual role has been found for upd3 expression, linked to increased/decreased mycobacterial load at the beginning and later in infection, respectively. Finally, impl2 expression has been related to increased resistance in non-actively mating males. These results allow further assessment on the differences between sexes and highlights the role of the reproductive status in D. melanogaster to face infections, demonstrating their importance to determine resistance and tolerance against M. marinum infection.
Collapse
Affiliation(s)
- Marta Arch
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
| | - Maria Vidal
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Esther Fuentes
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
| | - Anmaw Shite Abat
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Department of Veterinary Paraclinical Studies, University of Gondar, Gondar, Ethiopia
| | - Pere-Joan Cardona
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
19
|
Abstract
Endocrine signaling networks control diverse biological processes and life history traits across metazoans. In both invertebrate and vertebrate taxa, steroid hormones regulate immune system function in response to intrinsic and environmental stimuli, such as microbial infection. The mechanisms of this endocrine-immune regulation are complex and constitute an ongoing research endeavor facilitated by genetically tractable animal models. The 20-hydroxyecdysone (20E) is the major steroid hormone in arthropods, primarily studied for its essential role in mediating developmental transitions and metamorphosis; 20E also modulates innate immunity in a variety of insect taxa. This review provides an overview of our current understanding of 20E-mediated innate immune responses. The prevalence of correlations between 20E-driven developmental transitions and innate immune activation are summarized across a range of holometabolous insects. Subsequent discussion focuses on studies conducted using the extensive genetic resources available in Drosophila that have begun to reveal the mechanisms underlying 20E regulation of immunity in the contexts of both development and bacterial infection. Lastly, I propose directions for future research into 20E regulation of immunity that will advance our knowledge of how interactive endocrine networks coordinate animals' physiological responses to environmental microbes.
Collapse
Affiliation(s)
- Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
20
|
Kasuya J, Johnson W, Chen HL, Kitamoto T. Dietary Supplementation with Milk Lipids Leads to Suppression of Developmental and Behavioral Phenotypes of Hyperexcitable Drosophila Mutants. Neuroscience 2023; 520:1-17. [PMID: 37004908 PMCID: PMC10200772 DOI: 10.1016/j.neuroscience.2023.03.027] [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: 10/01/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Dietary modifications often have a profound impact on the penetrance and expressivity of neurological phenotypes that are caused by genetic defects. Our previous studies in Drosophila melanogaster revealed that seizure-like phenotypes of gain-of-function voltage-gated sodium (Nav) channel mutants (paraShu, parabss1, and paraGEFS+), as well as other seizure-prone "bang-sensitive" mutants (eas and sda), were drastically suppressed by supplementation of a standard diet with milk whey. In the current study we sought to determine which components of milk whey are responsible for the diet-dependent suppression of their hyperexcitable phenotypes. Our systematic analysis reveals that supplementing the diet with a modest amount of milk lipids (0.26% w/v) mimics the effects of milk whey. We further found that a minor milk lipid component, α-linolenic acid, contributed to the diet-dependent suppression of adult paraShu phenotypes. Given that lipid supplementation during the larval stages effectively suppressed adult paraShu phenotypes, dietary lipids likely modify neural development to compensate for the defects caused by the mutations. Consistent with this notion, lipid feeding fully rescued abnormal dendrite development of class IV sensory neurons in paraShu larvae. Overall, our findings demonstrate that milk lipids are sufficient to ameliorate hyperexcitable phenotypes in Drosophila mutants, providing a foundation for future investigation of the molecular and cellular mechanisms by which dietary lipids modify genetically induced abnormalities in neural development, physiology, and behavior.
Collapse
Affiliation(s)
- Junko Kasuya
- Department of Anesthesia, Carver College of Medicine, University of Iowa, 1-376 BSB, 51 Newton Road, Iowa City, IA 52242, United States.
| | - Wayne Johnson
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, United States; Interdisciplinary Graduate Program in Genetics, University of Iowa, IA 52242, United States.
| | - Hung-Lin Chen
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA 52242, United States
| | - Toshihiro Kitamoto
- Interdisciplinary Graduate Program in Genetics, University of Iowa, IA 52242, United States.
| |
Collapse
|
21
|
Chen Y, Gong H, Tang D, Yu L, Long S, Zheng B, Luo D, Cai A. Liver proteomic analysis reveals the key proteins involved in host immune response to sepsis. PeerJ 2023; 11:e15294. [PMID: 37255592 PMCID: PMC10226476 DOI: 10.7717/peerj.15294] [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: 11/18/2022] [Accepted: 04/04/2023] [Indexed: 06/01/2023] Open
Abstract
Background Sepsis is a serious infection-induced response in the host, which can result in life-threatening organ dysfunction. It is of great importance to unravel the relationship between sepsis and host immune response and its mechanisms of action. Liver is one of the most vulnerable organs in sepsis, however, the specific pathogenesis of septic liver injury has not been well understood at the protein level. Methods A total of 12 healthy Sprague-Dawley (SD) male rats aged from 6 to 8 weeks were adaptively housed in individual cages in the specific pathogen free animal room. These lab rats were grouped into two groups: treatment (N = 9) and control (N = 3) groups; only three mice from the treatment group survived and were used for subsequent experiments. A TMT-based proteomic analysis for liver tissue was performed in the septic rat model. Results A total of 37,012 unique peptides were identified, and then 6,166 proteins were determined, among which 5,701 were quantifiable. Compared to the healthy control group, the septic rat group exhibited 162 upregulated and 103 downregulated differentially expressed proteins (DEPs). The upregulated and downregulated DEPs were the most significantly enriched into the complement and coagulation cascades and metabolic pathways. Protein-protein interaction (PPI) analysis further revealed that the upregulated and downregulated DEPs each clustered in a PPI network. Several highly connected upregulated and downregulated DEPs were also enriched into the complement and coagulation cascades pathways and metabolic pathways, respectively. The parallel reaction monitoring (PRM) results of the selected DEPs were consistent with the results of the TMT analysis, supporting the proteomic data. Conclusion Our findings highlight the roles of complement and coagulation cascades and metabolic pathways that may play vital roles in the host immune response. The DEPs may serve as clinically potential treatment targets for septic liver injury.
Collapse
Affiliation(s)
- Yingying Chen
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Hui Gong
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Donge Tang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| | - Lan Yu
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Shoubin Long
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Bao Zheng
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Dixian Luo
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Anji Cai
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
22
|
Zhang R, Chen X, Wang Y, Bai X, Yang Q, Zhong Y, Yu XQ, Jin F, Yang W. BmMD-2A responds to 20-hydroxyecdysone and regulates Bombyx mori silkworm innate immunity in larva-to-pupa metamorphosis. INSECT SCIENCE 2023; 30:411-424. [PMID: 35871306 DOI: 10.1111/1744-7917.13097] [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: 02/18/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
20E-hydroxyecdysone (20E) plays important roles in larval molting and metamorphosis in insects and is also involved in the insect innate immune response. Insect metamorphosis is a highly successful strategy for environmental adaptation and is the most vulnerable stage during which the insect is susceptible to various pathogens. 20E regulates a series of antimicrobial peptides (AMPs) through the immunodeficiency (IMD) pathway activation in Drosophila; nevertheless, whether other immune pathways are involved in 20E-regulated insect immunity is unknown. Our previous studies showed that BmMD-2A is a member of the MD-2-related lipid recognition (ML) family of proteins that are involved in the Bombyx mori innate immunity Toll signaling pathway. In this study, we further demonstrate that BmMD-2A is also positively regulated by 20E, and the BmMD-2A neutralization experiment suggested that 20E activates some downstream immune effect factors, the AMP genes against Escherichia coli and Staphylococcus aureus, through the regulation of BmMD-2A in larval metamorphosis, implying that B. mori may use the Toll-ML signaling pathway to maintain innate immune balance in the larval-pupal metamorphosis stage, which is a different innate immunity pathway regulated by 20E compared to the IMD pathway in Drosophila.
Collapse
Affiliation(s)
- Ruonan Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory of Bio-Pesticide Creation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Xue Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuan Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xu Bai
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qiong Yang
- The Sericultural and Agri-Food Research Institute of the Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yangjin Zhong
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Fengliang Jin
- Guangdong Laboratory of Bio-Pesticide Creation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Wanying Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| |
Collapse
|
23
|
Okamoto N, Fujinaga D, Yamanaka N. Steroid hormone signaling: What we can learn from insect models. VITAMINS AND HORMONES 2023; 123:525-554. [PMID: 37717997 DOI: 10.1016/bs.vh.2022.12.006] [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] [Indexed: 02/09/2023]
Abstract
Ecdysteroids are a group of steroid hormones in arthropods with pleiotropic functions throughout their life history. Ecdysteroid research in insects has made a significant contribution to our current understanding of steroid hormone signaling in metazoans, but how far can we extrapolate our findings in insects to other systems, such as mammals? In this chapter, we compare steroid hormone signaling in insects and mammals from multiple perspectives and discuss similarities and differences between the two lineages. We also highlight a few understudied areas and remaining questions of steroid hormone biology in metazoans and propose potential future research directions.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Daiki Fujinaga
- Department of Entomology, University of California, Riverside, CA, United States
| | - Naoki Yamanaka
- Department of Entomology, University of California, Riverside, CA, United States.
| |
Collapse
|
24
|
Ning L, Wang X, Xuan B, Ma Y, Yan Y, Gao Z, Tong T, Cui Z, Chen H, Li X, Hong J, Wang Z. Identification and investigation of depression-related molecular subtypes in inflammatory bowel disease and the anti-inflammatory mechanisms of paroxetine. Front Immunol 2023; 14:1145070. [PMID: 36923403 PMCID: PMC10008943 DOI: 10.3389/fimmu.2023.1145070] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
Background Up to 40 per cent of people with active inflammatory bowel disease (IBD) also suffer from mood disorders such as anxiety and depression. Notwithstanding, the fundamental biological pathways driving depression in IBD remain unknown. Methods We identified 33 core genes that drive depression in IBD patients and performed consensus molecular subtyping with the NMF algorithm in IBD. The CIBERSORT were employed to quantify the immune cells. Metabolic signature was characterized using the "IOBR" R package. The scoring system (D. score) based on PCA. Pre-clinical models are constructed using DSS. Results Using transcriptome data from the GEO database of 630 IBD patients, we performed a thorough analysis of the correlation between IBD and depression in this research. Firstly, the samples were separated into two different molecular subtypes (D. cluster1 and D. cluster2) based on their biological signatures. Moreover, the immunological and metabolic differences between them were evaluated, and we discovered that D. cluster2 most closely resembled IBD patients concomitant with depression. We also developed a scoring system to assess the IBD-related depression and predict clinical response to anti-TNF- therapy, with a higher D. score suggesting more inflammation and worse reaction to biological therapies. Ultimately, we also identified through animal experiments an antidepressant, paroxetine, has the added benefit of lowering intestinal inflammation by controlling microorganisms in the digestive tract. Conclusions This study highlights that IBD patients with or without depression show significant variations and antidepressant paroxetine may help reduce intestinal inflammation.
Collapse
Affiliation(s)
- Lijun Ning
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyuan Wang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanru Ma
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqing Yan
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyun Gao
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianying Tong
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Cui
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobo Li
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhua Wang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Shanghai Cancer Institute, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Magnotti F, Chirita D, Dalmon S, Martin A, Bronnec P, Sousa J, Helynck O, Lee W, Kastner DL, Chae JJ, McDermott MF, Belot A, Popoff M, Sève P, Georgin-Lavialle S, Munier-Lehmann H, Tran TA, De Langhe E, Wouters C, Jamilloux Y, Henry T. Steroid hormone catabolites activate the pyrin inflammasome through a non-canonical mechanism. Cell Rep 2022; 41:111472. [PMID: 36223753 PMCID: PMC9626387 DOI: 10.1016/j.celrep.2022.111472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 06/18/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
The pyrin inflammasome acts as a guard of RhoA GTPases and is central to immune defenses against RhoA-manipulating pathogens. Pyrin activation proceeds in two steps. Yet, the second step is still poorly understood. Using cells constitutively activated for the pyrin step 1, a chemical screen identifies etiocholanolone and pregnanolone, two catabolites of testosterone and progesterone, acting at low concentrations as specific step 2 activators. High concentrations of these metabolites fully and rapidly activate pyrin, in a human specific, B30.2 domain-dependent manner and without inhibiting RhoA. Mutations in MEFV, encoding pyrin, cause two distinct autoinflammatory diseases pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND) and familial Mediterranean fever (FMF). Monocytes from PAAND patients, and to a lower extent from FMF patients, display increased responses to these metabolites. This study identifies an unconventional pyrin activation mechanism, indicates that endogenous steroid catabolites can drive autoinflammation, through the pyrin inflammasome, and explains the "steroid fever" described in the late 1950s upon steroid injection in humans.
Collapse
Affiliation(s)
- Flora Magnotti
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Daria Chirita
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Sarah Dalmon
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Amandine Martin
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Pauline Bronnec
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Jeremy Sousa
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France
| | - Olivier Helynck
- Institut Pasteur, Université de Paris Cité, CNRS UMR3523, Chemistry and Biocatalysis Unit, 75724 Paris Cedex 15, France
| | - Wonyong Lee
- Inflammatory Disease Section, Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Jae Jin Chae
- Inflammatory Disease Section, Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK
| | - Alexandre Belot
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France; Department of Pediatric Nephrology, Rheumatology, Dermatology, Reference Centre for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Femme Mère Enfant, CHU Lyon, Lyon, France; LIFE, Lyon Immunopathology Federation, Lyon, France
| | | | - Pascal Sève
- Department of Internal Medicine, University Hospital Croix-Rousse, Lyon 1 University, Lyon, France
| | - Sophie Georgin-Lavialle
- Sorbonne University, Department of Internal Medicine, Tenon Hospital, DMU 3ID, AP-HP, National Reference Center for Autoinflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA), INSERM U938, Paris, France
| | - Hélène Munier-Lehmann
- Institut Pasteur, Université de Paris Cité, CNRS UMR3523, Chemistry and Biocatalysis Unit, 75724 Paris Cedex 15, France
| | - Tu Anh Tran
- Department of Pediatrics, Carémeau Hospital, CHU Nîmes, Nîmes, France
| | - Ellen De Langhe
- Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Tissue Homeostasis and Disease, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Laboratory of Adaptive Immunology & Immunobiology, Leuven, Belgium; Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yvan Jamilloux
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France; LIFE, Lyon Immunopathology Federation, Lyon, France; Department of Internal Medicine, University Hospital Croix-Rousse, Lyon 1 University, Lyon, France.
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, University Lyon, 69007 Lyon, France.
| |
Collapse
|
26
|
Zhou Q, Zhu X, Li Y, Yang P, Wang S, Ning K, Chen S. Intestinal microbiome-mediated resistance against vibriosis for Cynoglossus semilaevis. MICROBIOME 2022; 10:153. [PMID: 36138436 PMCID: PMC9503257 DOI: 10.1186/s40168-022-01346-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/10/2022] [Indexed: 06/06/2023]
Abstract
BACKGROUND Infectious diseases have caused huge economic loss and food security issues in fish aquaculture. Current management and breeding strategies heavily rely on the knowledge of regulative mechanisms underlying disease resistance. Though the intestinal microbial community was linked with disease infection, there is little knowledge about the roles of intestinal microbes in fish disease resistance. Cynoglossus semilaevis is an economically important and widely cultivated flatfish species in China. However, it suffers from outbreaks of vibriosis, which results in huge mortalities and economic loss. RESULTS Here, we used C. semilaevis as a research model to investigate the host-microbiome interactions in regulating vibriosis resistance. The resistance to vibriosis was reflected in intestinal microbiome on both taxonomic and functional levels. Such differences also influenced the host gene expressions in the resistant family. Moreover, the intestinal microbiome might control the host immunological homeostasis and inflammation to enhance vibriosis resistance through the microbe-intestine-immunity axis. For example, Phaeobacter regulated its hdhA gene and host cyp27a1 gene up-expressed in bile acid biosynthesis pathways, but regulated its trxA gene and host akt gene down-expressed in proinflammatory cytokines biosynthesis pathways, to reduce inflammation and resist disease infection in the resistant family. Furthermore, the combination of intestinal microbes and host genes as biomarkers could accurately differentiate resistant family from susceptible family. CONCLUSION Our study uncovered the regulatory patterns of the microbe-intestine-immunity axis that may contribute to vibriosis resistance in C. semilaevis. These findings could facilitate the disease control and selective breeding of superior germplasm with high disease resistance in fish aquaculture. Video Abstract.
Collapse
Affiliation(s)
- Qian Zhou
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences/Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture; Shandong Key Laboratory for Marine Fishery Biotechnology and Genetic Breeding; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071, Shandong, China
| | - Xue Zhu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Yangzhen Li
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences/Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture; Shandong Key Laboratory for Marine Fishery Biotechnology and Genetic Breeding; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071, Shandong, China
| | - Pengshuo Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Shengpeng Wang
- Dezhou Key Laboratory for Applied Bile Acid Research, Shandong Longchang Animal Health Product Co., Ltd., Qihe, Shandong Lachance Co., Ltd., Jinan, 251100, Shandong, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center of AI Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
| | - Songlin Chen
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences/Key Laboratory for Sustainable Development of Marine Fisheries, Ministry of Agriculture; Shandong Key Laboratory for Marine Fishery Biotechnology and Genetic Breeding; Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266071, Shandong, China.
| |
Collapse
|
27
|
Yu S, Luo F, Xu Y, Zhang Y, Jin LH. Drosophila Innate Immunity Involves Multiple Signaling Pathways and Coordinated Communication Between Different Tissues. Front Immunol 2022; 13:905370. [PMID: 35911716 PMCID: PMC9336466 DOI: 10.3389/fimmu.2022.905370] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune response provides the first line of defense against invading pathogens, and immune disorders cause a variety of diseases. The fruit fly Drosophila melanogaster employs multiple innate immune reactions to resist infection. First, epithelial tissues function as physical barriers to prevent pathogen invasion. In addition, macrophage-like plasmatocytes eliminate intruders through phagocytosis, and lamellocytes encapsulate large particles, such as wasp eggs, that cannot be phagocytosed. Regarding humoral immune responses, the fat body, equivalent to the mammalian liver, secretes antimicrobial peptides into hemolymph, killing bacteria and fungi. Drosophila has been shown to be a powerful in vivo model for studying the mechanism of innate immunity and host-pathogen interactions because Drosophila and higher organisms share conserved signaling pathways and factors. Moreover, the ease with which Drosophila genetic and physiological characteristics can be manipulated prevents interference by adaptive immunity. In this review, we discuss the signaling pathways activated in Drosophila innate immunity, namely, the Toll, Imd, JNK, JAK/STAT pathways, and other factors, as well as relevant regulatory networks. We also review the mechanisms by which different tissues, including hemocytes, the fat body, the lymph gland, muscles, the gut and the brain coordinate innate immune responses. Furthermore, the latest studies in this field are outlined in this review. In summary, understanding the mechanism underlying innate immunity orchestration in Drosophila will help us better study human innate immunity-related diseases.
Collapse
|
28
|
Martinson EO, Chen K, Valzania L, Brown MR, Strand MR. Insulin-like peptide 3 stimulates hemocytes to proliferate in anautogenous and facultatively autogenous mosquitoes. J Exp Biol 2022; 225:274275. [PMID: 35129195 PMCID: PMC8976944 DOI: 10.1242/jeb.243460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022]
Abstract
Most mosquito species are anautogenous, which means they must blood feed on a vertebrate host to produce eggs, while a few are autogenous and can produce eggs without blood feeding. Egg formation is best understood in the anautogenous mosquito Aedes aegypti, where insulin-like peptides (ILPs), ovary ecdysteroidogenic hormone (OEH) and 20-hydroxyecdysone (20E) interact to regulate gonadotrophic cycles. Circulating hemocytes also approximately double in abundance in conjunction with a gonadotrophic cycle, but the factors responsible for stimulating this increase remain unclear. Focusing on Ae. aegypti, we determined that hemocyte abundance similarly increased in intact blood-fed females and decapitated blood-fed females that were injected with ILP3, whereas OEH, 20E or heat-killed bacteria had no stimulatory activity. ILP3 upregulated insulin-insulin growth factor signaling in hemocytes, but few genes - including almost no transcripts for immune factors - were differentially expressed. ILP3 also stimulated circulating hemocytes to increase in two other anautogenous (Anopheles gambiae and Culex quinquefasciatus) and two facultatively autogenous mosquitoes (Aedes atropalpus and Culex pipiens molestus), but had no stimulatory activity in the obligately autogenous mosquito Toxorhynchites amboinensis. Altogether, our results identify ILPs as the primary regulators of hemocyte proliferation in association with egg formation, but also suggest this response has been lost in the evolution of obligate autogeny.
Collapse
Affiliation(s)
- Ellen O Martinson
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Kangkang Chen
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Luca Valzania
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Mark R Brown
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
29
|
Nunes C, Koyama T, Sucena É. Co-option of immune effectors by the hormonal signalling system triggering metamorphosis in Drosophila melanogaster. PLoS Genet 2021; 17:e1009916. [PMID: 34843450 PMCID: PMC8659296 DOI: 10.1371/journal.pgen.1009916] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/09/2021] [Accepted: 10/29/2021] [Indexed: 11/24/2022] Open
Abstract
Insect metamorphosis is triggered by the production, secretion and degradation of 20-hydroxyecdysone (ecdysone). In addition to its role in developmental regulation, increasing evidence suggests that ecdysone is involved in innate immunity processes, such as phagocytosis and the induction of antimicrobial peptide (AMP) production. AMP regulation includes systemic responses as well as local responses at surface epithelia that contact with the external environment. At pupariation, Drosophila melanogaster increases dramatically the expression of three AMP genes, drosomycin (drs), drosomycin-like 2 (drsl2) and drosomycin-like 5 (drsl5). We show that the systemic action of drs at pupariation is dependent on ecdysone signalling in the fat body and operates via the ecdysone downstream target, Broad. In parallel, ecdysone also regulates local responses, specifically through the activation of drsl2 expression in the gut. Finally, we confirm the relevance of this ecdysone dependent AMP expression for the control of bacterial load by showing that flies lacking drs expression in the fat body have higher bacterial persistence over metamorphosis. In contrast, local responses may be redundant with the systemic effect of drs since reduction of ecdysone signalling or of drsl2 expression has no measurable negative effect on bacterial load control in the pupa. Together, our data emphasize the importance of the association between ecdysone signalling and immunity using in vivo studies and establish a new role for ecdysone at pupariation, which impacts developmental success by regulating the immune system in a stage-dependent manner. We speculate that this co-option of immune effectors by the hormonal system may constitute an anticipatory mechanism to control bacterial numbers in the pupa, at the core of metamorphosis evolution.
Collapse
Affiliation(s)
- Catarina Nunes
- Evolution and Development Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Takashi Koyama
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Élio Sucena
- Evolution and Development Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
30
|
Wang GJ, Wang WW, Liu Y, Chai LQ, Wang GX, Liu XS, Wang YF, Wang JL. Steroid hormone 20-hydroxyecdysone promotes CTL1-mediated cellular immunity in Helicoverpa armigera. INSECT SCIENCE 2021; 28:1399-1413. [PMID: 32677271 DOI: 10.1111/1744-7917.12851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 06/11/2023]
Abstract
Mermithid nematodes, such as Ovomermis sinensis, are used as biological control agents against many insect pests, including cotton bollworm (Helicoverpa armigera). However, given the host's robust immune system, the infection rate of O. sinensis is low, thus restricting its widespread use. To understand the host defense mechanisms against mermithid nematodes, we identified and characterized a protein involved in the recognition of O. sinensis, the potential O. sinensis-binding protein C-type lectin 1 (HaCTL1a and/or HaCTL1b), which was eluted from the surface of O. sinensis after incubation with H. armigera plasma. HaCTL1b is homologous to the previously reported HaCTL1a protein. HaCTL1 was predominantly expressed in hemocytes and was induced by the steroid hormone 20-hydroxyecdysone through ecdysone receptor (HaEcR) or ultraspiracle (HaUSP), or both. Binding assays confirmed the interactions of the HaCTL1 proteins with O. sinensis but not with Romanomermis wuchangensis, a parasitic nematode of mosquito. Moreover, the HaCTL1 proteins were secreted into the hemocoel and promoted hemocyte-mediated encapsulation and phagocytosis. A knockdown of HaEcR and/or HaUSP resulted in compromised encapsulation and phagocytosis. Thus, HaCTL1 appears to modulate cellular immunity in the defense against parasitic nematodes, and the 20-hydroxyecdysone-HaEcR-HaUSP complex is involved in regulating the process.
Collapse
Affiliation(s)
- Gui-Jie Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Wen-Wen Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Yu Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Lian-Qin Chai
- School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Guo-Xiu Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Xu-Sheng Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Yu-Feng Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Jia-Lin Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
31
|
Guzman RM, Howard ZP, Liu Z, Oliveira RD, Massa AT, Omsland A, White SN, Goodman AG. Natural genetic variation in Drosophila melanogaster reveals genes associated with Coxiella burnetii infection. Genetics 2021; 217:6117219. [PMID: 33789347 PMCID: PMC8045698 DOI: 10.1093/genetics/iyab005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022] Open
Abstract
The gram-negative bacterium Coxiella burnetii is the causative agent of Query (Q) fever in humans and coxiellosis in livestock. Host genetics are associated with C. burnetii pathogenesis both in humans and animals; however, it remains unknown if specific genes are associated with severity of infection. We employed the Drosophila Genetics Reference Panel to perform a genome-wide association study to identify host genetic variants that affect host survival to C. burnetii infection. The genome-wide association study identified 64 unique variants (P < 10−5) associated with 25 candidate genes. We examined the role each candidate gene contributes to host survival during C. burnetii infection using flies carrying a null mutation or RNAi knockdown of each candidate. We validated 15 of the 25 candidate genes using at least one method. This is the first report establishing involvement of many of these genes or their homologs with C. burnetii susceptibility in any system. Among the validated genes, FER and tara play roles in the JAK/STAT, JNK, and decapentaplegic/TGF-β signaling pathways which are components of known innate immune responses to C. burnetii infection. CG42673 and DIP-ε play roles in bacterial infection and synaptic signaling but have no previous association with C. burnetii pathogenesis. Furthermore, since the mammalian ortholog of CG13404 (PLGRKT) is an important regulator of macrophage function, CG13404 could play a role in host susceptibility to C. burnetii through hemocyte regulation. These insights provide a foundation for further investigation regarding the genetics of C. burnetii susceptibility across a wide variety of hosts.
Collapse
Affiliation(s)
- Rosa M Guzman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Zachary P Howard
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ziying Liu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ryan D Oliveira
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Alisha T Massa
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Anders Omsland
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Stephen N White
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.,USDA-ARS Animal Disease Research, Pullman, WA 99164, USA.,Center for Reproductive Biology, Washington State University, Pullman, WA 99164, USA
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.,Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
32
|
Banisch TU, Slaidina M, Gupta S, Ho M, Gilboa L, Lehmann R. A transitory signaling center controls timing of primordial germ cell differentiation. Dev Cell 2021; 56:1742-1755.e4. [PMID: 34081907 PMCID: PMC8330407 DOI: 10.1016/j.devcel.2021.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/07/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Organogenesis requires exquisite spatiotemporal coordination of cell morphogenesis, migration, proliferation, and differentiation of multiple cell types. For gonads, this involves complex interactions between somatic and germline tissues. During Drosophila ovary morphogenesis, primordial germ cells (PGCs) either are sequestered in stem cell niches and are maintained in an undifferentiated germline stem cell state or transition directly toward differentiation. Here, we identify a mechanism that links hormonal triggers of somatic tissue morphogenesis with PGC differentiation. An early ecdysone pulse initiates somatic swarm cell (SwC) migration, positioning these cells close to PGCs. A second hormone peak activates Torso-like signal in SwCs, which stimulates the Torso receptor tyrosine kinase (RTK) signaling pathway in PGCs promoting their differentiation by de-repression of the differentiation gene, bag of marbles. Thus, systemic temporal cues generate a transitory signaling center that coordinates ovarian morphogenesis with stem cell self-renewal and differentiation programs, highlighting a more general role for such centers in reproductive and developmental biology.
Collapse
Affiliation(s)
- Torsten U Banisch
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| | - Maija Slaidina
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Selena Gupta
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Megan Ho
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ruth Lehmann
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
33
|
Coates JA, Brooks E, Brittle AL, Armitage EL, Zeidler MP, Evans IR. Identification of functionally distinct macrophage subpopulations in Drosophila. eLife 2021; 10:e58686. [PMID: 33885361 PMCID: PMC8062135 DOI: 10.7554/elife.58686] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Vertebrate macrophages are a highly heterogeneous cell population, but while Drosophila blood is dominated by a macrophage-like lineage (plasmatocytes), until very recently these cells were considered to represent a homogeneous population. Here, we present our identification of enhancer elements labelling plasmatocyte subpopulations, which vary in abundance across development. These subpopulations exhibit functional differences compared to the overall population, including more potent injury responses and differential localisation and dynamics in pupae and adults. Our enhancer analysis identified candidate genes regulating plasmatocyte behaviour: pan-plasmatocyte expression of one such gene (Calnexin14D) improves wound responses, causing the overall population to resemble more closely the subpopulation marked by the Calnexin14D-associated enhancer. Finally, we show that exposure to increased levels of apoptotic cell death modulates subpopulation cell numbers. Taken together this demonstrates macrophage heterogeneity in Drosophila, identifies mechanisms involved in subpopulation specification and function and facilitates the use of Drosophila to study macrophage heterogeneity in vivo.
Collapse
Affiliation(s)
- Jonathon Alexis Coates
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Elliot Brooks
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Amy Louise Brittle
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Emma Louise Armitage
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Martin Peter Zeidler
- Department of Biomedical Science and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| | - Iwan Robert Evans
- Department of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
34
|
Mase A, Augsburger J, Brückner K. Macrophages and Their Organ Locations Shape Each Other in Development and Homeostasis - A Drosophila Perspective. Front Cell Dev Biol 2021; 9:630272. [PMID: 33777939 PMCID: PMC7991785 DOI: 10.3389/fcell.2021.630272] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
Across the animal kingdom, macrophages are known for their functions in innate immunity, but they also play key roles in development and homeostasis. Recent insights from single cell profiling and other approaches in the invertebrate model organism Drosophila melanogaster reveal substantial diversity among Drosophila macrophages (plasmatocytes). Together with vertebrate studies that show genuine expression signatures of macrophages based on their organ microenvironments, it is expected that Drosophila macrophage functional diversity is shaped by their anatomical locations and systemic conditions. In vivo evidence for diverse macrophage functions has already been well established by Drosophila genetics: Drosophila macrophages play key roles in various aspects of development and organogenesis, including embryogenesis and development of the nervous, digestive, and reproductive systems. Macrophages further maintain homeostasis in various organ systems and promote regeneration following organ damage and injury. The interdependence and interplay of tissues and their local macrophage populations in Drosophila have implications for understanding principles of organ development and homeostasis in a wide range of species.
Collapse
Affiliation(s)
- Anjeli Mase
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Jordan Augsburger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
| | - Katja Brückner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
35
|
Ray R, Potts R, Pietri JE. The Persistence of Escherichia coli Infection in German Cockroaches (Blattodea: Blattellidae) Varies Between Host Developmental Stages and is Influenced by the Gut Microbiota. JOURNAL OF MEDICAL ENTOMOLOGY 2020; 57:1964-1971. [PMID: 32516418 DOI: 10.1093/jme/tjaa108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 06/11/2023]
Abstract
The German cockroach, Blatella germanica (L.), is a suspected vector of several enteric bacterial pathogens, including Escherichia coli, among livestock and humans. However, little is known about the factors that influence bacterial transmission by cockroaches. Here, we orally infected B. germanica with various laboratory and field strains of E. coli and examined bacterial titers over time to shed new light on the factors that influence the dynamics of infection. Our results reveal that a laboratory strain of E. coli is largely cleared within 48 h while one field isolate can persist in a majority of cockroaches (80-100%) for longer than 3 d with minimal impact on cockroach longevity. We also find that the ability to clear some strains of E. coli is greater in cockroach nymphs than adults. Notably, no differential expression of the antimicrobial gene lysozyme was observed between nymphs and adults or in infected groups. However, clearance of E. coli was significantly reduced in gnotobiotic cockroaches that were reared in the absence of environmental bacteria, suggesting a protective role for the microbiota against exogenous bacterial pathogens. Together, these results demonstrate that the interactions between cockroaches and enteric bacterial pathogens are highly dynamic and influenced by a combination of microbial, host, and environmental parameters. Such factors may affect the disease transmission capacity of cockroaches in nature and should be further considered in both lab and field studies.
Collapse
Affiliation(s)
- Ritesh Ray
- Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion, SD
| | - Rashaun Potts
- Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion, SD
| | | |
Collapse
|
36
|
Nunes C, Sucena É, Koyama T. Endocrine regulation of immunity in insects. FEBS J 2020; 288:3928-3947. [PMID: 33021015 DOI: 10.1111/febs.15581] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Organisms have constant contact with potentially harmful agents that can compromise their fitness. However, most of the times these agents fail to cause serious disease by virtue of the rapid and efficient immune responses elicited in the host that can range from behavioural adaptations to immune system triggering. The immune system of insects does not comprise the adaptive arm, making it less complex than that of vertebrates, but key aspects of the activation and regulation of innate immunity are conserved across different phyla. This is the case for the hormonal regulation of immunity as a part of the broad organismal responses to external conditions under different internal states. In insects, depending on the physiological circumstances, distinct hormones either enhance or suppress the immune response integrating individual (and often collective) responses physiologically and behaviourally. In this review, we provide an overview of our current knowledge on the endocrine regulation of immunity in insects, its mechanisms and implications on metabolic adaptation and behaviour. We highlight the importance of this multilayered regulation of immunity in survival and reproduction (fitness) and its dependence on the hormonal integration with other mechanisms and life-history traits.
Collapse
Affiliation(s)
| | - Élio Sucena
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Huang S, Yi Q, Lian X, Xu S, Yang C, Sun J, Wang L, Song L. The involvement of ecdysone and ecdysone receptor in regulating the expression of antimicrobial peptides in Chinese mitten crab, Eriocheir sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 111:103757. [PMID: 32485180 DOI: 10.1016/j.dci.2020.103757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
The ecdysone, 20-hydroxyecdysone (20E) and ecdysone receptor (EcR), are regarded as the key regulators of development, metamorphosis, and growth in arthropods. In the present study, the role of 20E and EsEcR in regulating the expression of antimicrobial peptides (AMPs) was investigated in Chinese mitten crab, Eriocheir sinensis. The concentration of 20E in plasma was significantly (p < 0.05) up-regulated from 3 h to 12 h after lipopolysaccharide (LPS) stimulation. The mRNA expression level of EsEcR-4 in hemocytes was significantly (p < 0.01) up-regulated from 6 h to 24 h after LPS stimulation, while no significant changes of EsEcR-2 and EsEcR-3 transcripts were observed. After 20E injection, EsEcR-4 expression level was significantly increased from 12 h to 48 h with the highest level at 24 h (4.34-fold compared to the control group, p < 0.01), and the mRNA expression levels of AMPs (EsALF-2, EsLYZ and EsCrus) in hemocytes were significantly increased from 6 h to 24 h with the peak level of 2.93-fold (p < 0.01), 2.33-fold (p < 0.01) and 2.75-fold (p < 0.01) at 12 h, respectively. After EsEcR-4 expression was interfered with specific dsRNA, a significant reduction of EsALF-2 (0.56-fold compared to the control group, p < 0.01), EsLYZ (0.27-fold, p < 0.01) and EsCrus (0.41-fold, p < 0.01) mRNA expression level was observed in dsEsEcR-4+LPS group at 12 h post LPS stimulation. Moreover, the mRNA expression levels of EsDorsal and EsJNK in hemocytes were significantly (p < 0.05) increased from 6 h to 24 h post 20E injection, and the phosphorylation of Dorsal and JNK in the hemocytes were significantly (p < 0.01) up-regulated at 3 h post 20E injection, while that in dsEsEcR-4+LPS group were significantly decreased after LPS stimulation compared to dsEsEGFP+LPS group. Taken together, these results suggested that 20E and EsEcR-4 play important roles in regulating the expression level of AMPs in the immune responses of E. sinensis by regulating the mRNA expression level and phosphorylation of Dorsal and JNK.
Collapse
Affiliation(s)
- Shu Huang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Qilin Yi
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Xingye Lian
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Siqi Xu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Chuanyan Yang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
38
|
Li M, Chen Z, Qian J, Wei F, Zhang G, Wang Y, Wei G, Hu Z, Dong L, Chen S. Composition and function of rhizosphere microbiome of Panax notoginseng with discrepant yields. Chin Med 2020; 15:85. [PMID: 32793300 PMCID: PMC7418314 DOI: 10.1186/s13020-020-00364-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/03/2020] [Indexed: 11/21/2022] Open
Abstract
Background Panax notoginseng is a highly valuable medicinal plant. Reduced P. notoginseng yield is a common and serious problem that arises in a continuous cropping system. Variation in the composition and function of soil microbial community is considered the primary cause of yield reduction. Methods This study used shotgun metagenomic sequencing approaches to describe the taxonomic and functional features of P. notoginseng rhizosphere microbiome and screen microbial taxa and functional traits related to yields. Results At the family and genus level, a total of 43 families and 45 genera (relative abundance > 0.1%) were obtained, and the correlation with the yield of P. notoginseng was further analyzed. Nitrosomonadaceae, Xanthomonadaceae, Mycobacterium and Arthrobacter that were enriched in soils with higher yields were positively correlated with P. notoginseng yields, thereby suggesting that they might increase yields. Negative correlation coefficients indicated that Xanthobacteraceae, Caulobacteraceae, Oxalobacteraceae, Chitinophagaceae, Sphingomonas, Hyphomicrobium, Variovorax and Phenylobacterium might be detrimental to P. notoginseng growth. A total of 85 functional traits were significantly (P < 0.05) correlated with P. notoginseng yields. Functional traits, likely steroid biosynthesis and MAPK signaling pathway were positively correlated with P. notoginseng yields. In contrast, functional traits, such as bacterial secretion system, ABC transporters, metabolism of xenobiotics by cytochrome P450 and drug metabolism–cytochrome P450, were negatively associated with yields. Conclusions This study describes an overview of the rhizosphere microbiome of P. notoginseng with discrepant yields and identifies the taxa and functional traits related to yields. Our results provide valuable information to guide the isolation and culture of potentially beneficial microorganisms and to utilize the power of the microbiome to increase plant yields in a continuous cropping system.
Collapse
Affiliation(s)
- Mengzhi Li
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065 China.,Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Zhongjian Chen
- Institute of Sanqi Research, Wenshan University, Wenshan, 663000 China.,Wenshan Miaoxiang Notoginseng Technology, Co., Ltd, Wenshan, 663000 China
| | - Jun Qian
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China.,College of Pharmaceutical Science, Dali University, Dali, 671000 China
| | - Fugang Wei
- Wenshan Miaoxiang Notoginseng Technology, Co., Ltd, Wenshan, 663000 China
| | - Guozhuang Zhang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Yong Wang
- Institute of Sanqi Research, Wenshan University, Wenshan, 663000 China
| | - Guangfei Wei
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Zhigang Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065 China
| | - Linlin Dong
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| | - Shilin Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065 China.,Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, China Institute of Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, 100700 China
| |
Collapse
|
39
|
Bartolo G, Gonzalez LO, Alameh S, Valencia CA, Martchenko Shilman M. Identification of glucocorticoid receptor in Drosophila melanogaster. BMC Microbiol 2020; 20:161. [PMID: 32539689 PMCID: PMC7296755 DOI: 10.1186/s12866-020-01848-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Vertebrate glucocorticoid receptor (GR) is an evolutionary-conserved cortisol-regulated nuclear receptor that controls key metabolic and developmental pathways. Upon binding to cortisol, GR acts as an immunosuppressive transcription factor. Drosophila melanogaster, a model organism to study innate immunity, can also be immunosuppressed by glucocorticoids. However, while the genome of fruit fly harbors 18 nuclear receptor genes, the functional homolog of vertebrate GR has not been identified. RESULTS In this study, we demonstrated that while D. melanogaster is susceptible to Saccharomyces cerevisiae oral infection, the oral exposure to cortisol analogs, cortisone acetate or estrogen, increases fly sensitivity to yeast challenge. To understand the mechanism of this steroid-induced immunosuppression, we identified the closest genetic GR homolog as D. melanogaster Estrogen Related Receptor (ERR) gene. We discovered that Drosophila ERR is necessary for cortisone acetate- and estrogen-mediated increase in sensitivity to fungal infection: while ERR mutant flies are as sensitive to the fungal challenge as the wildtype flies, the yeast-sensitivity of ERR mutants is not increased by these steroids. Interestingly, the fungal cortisone analog, ergosterol, did not increase the susceptibility of Drosophila to yeast infection. The immunosuppressive effect of steroids on the sensitivity of flies to fungi is evolutionary conserved in insects, as we show that estrogen significantly increases the yeast-sensitivity of Culex quinquefasciatus mosquitoes, whose genome contains a close ortholog of the fly ERR gene. CONCLUSIONS This study identifies a D. melanogaster gene that structurally resembles vertebrate GR and is functionally necessary for the steroid-mediated immunosuppression to fungal infections.
Collapse
Affiliation(s)
- Gloria Bartolo
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Leandra O Gonzalez
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Saleem Alameh
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - C Alexander Valencia
- Aperiomics, Inc., Sterling, VA, 20166, USA
- Lake Erie College of Osteopathic Medicine, 1858 W Grandview Blvd, Erie, PA, 16509, USA
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | |
Collapse
|
40
|
Siriyappagouder P, Galindo-Villegas J, Dhanasiri AKS, Zhang Q, Mulero V, Kiron V, Fernandes JMO. Pseudozyma Priming Influences Expression of Genes Involved in Metabolic Pathways and Immunity in Zebrafish Larvae. Front Immunol 2020; 11:978. [PMID: 32528473 PMCID: PMC7256946 DOI: 10.3389/fimmu.2020.00978] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/24/2020] [Indexed: 12/14/2022] Open
Abstract
Fungi, particularly yeasts, are known essential components of the host microbiota but their functional relevance in development of immunity and physiological processes of fish remains to be elucidated. In this study, we used a transcriptomic approach and a germ-free (GF) fish model to determine the response of newly hatched zebrafish larvae after 24 h exposure to Pseudozyma sp. when compared to conventionally-raised (CR) larvae. We observed 59 differentially expressed genes in Pseudozyma-exposed GF zebrafish larvae compared to their naïve control siblings. Surprisingly, in CR larvae, there was not a clear transcriptome difference between Pseudozyma-exposed and control larvae. Differentially expressed genes in GF larvae were involved in host metabolic pathways, mainly peroxisome proliferator-activated receptors, steroid hormone biosynthesis, drug metabolism and bile acid biosynthesis. We also observed a significant change in the transcript levels of immune-related genes, namely complement component 3a, galectin 2b, ubiquitin specific peptidase 21, and aquaporins. Nevertheless, we did not observe any significant response at the cellular level, since there were no differences between neutrophil migration or proliferation between control and yeast-exposed GF larvae. Our findings reveal that exposure to Pseudozyma sp. may affect metabolic pathways and immune-related processes in germ-free zebrafish, suggesting that commensal yeast likely play a significant part in the early development of fish larvae.
Collapse
Affiliation(s)
| | - Jorge Galindo-Villegas
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
- Department of Cell Biology and Histology, Faculty of Biology, Institute of Biomedical Research of Murcia-Arrixaca, Campus Universitario de Espinardo, University of Murcia, Murcia, Spain
| | | | - Qirui Zhang
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, Institute of Biomedical Research of Murcia-Arrixaca, Campus Universitario de Espinardo, University of Murcia, Murcia, Spain
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | |
Collapse
|
41
|
Pondeville E, Puchot N, Parvy JP, Carissimo G, Poidevin M, Waterhouse RM, Marois E, Bourgouin C. Hemocyte-targeted gene expression in the female malaria mosquito using the hemolectin promoter from Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 120:103339. [PMID: 32105779 PMCID: PMC7181189 DOI: 10.1016/j.ibmb.2020.103339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Hemocytes, the immune cells in mosquitoes, participate in immune defenses against pathogens including malaria parasites. Mosquito hemocytes can also be infected by arthropod-borne viruses but the pro- or anti-viral nature of this interaction is unknown. Although there has been progress on hemocyte characterization during pathogen infection in mosquitoes, the specific contribution of hemocytes to immune responses and the hemocyte-specific functions of immune genes and pathways remain unresolved due to the lack of genetic tools to manipulate gene expression in these cells specifically. Here, we used the Gal4-UAS system to characterize the activity of the Drosophila hemocyte-specific hemolectin promoter in the adults of Anopheles gambiae, the malaria mosquito. We established an hml-Gal4 driver line that we further crossed to a fluorescent UAS responder line, and examined the expression pattern in the adult progeny driven by the hml promoter. We show that the hml regulatory region drives hemocyte-specific transgene expression in a subset of hemocytes, and that transgene expression is triggered after a blood meal. The hml promoter drives transgene expression in differentiating prohemocytes as well as in differentiated granulocytes. Analysis of different immune markers in hemocytes in which the hml promoter drives transgene expression revealed that this regulatory region could be used to study phagocytosis as well as melanization. Finally, the hml promoter drives transgene expression in hemocytes in which o'nyong-nyong virus replicates. Altogether, the Drosophila hml promoter constitutes a good tool to drive transgene expression in hemocyte only and to analyze the function of these cells and the genes they express during pathogen infection in Anopheles gambiae.
Collapse
Affiliation(s)
- Emilie Pondeville
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| | - Nicolas Puchot
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | | | - Guillaume Carissimo
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Mickael Poidevin
- Centre de Génétique Moléculaire, CNRS UPR 2167, Gif-sur-Yvette, France
| | - Robert M Waterhouse
- Department of Ecology and Evolution, Swiss Institute of Bioinformatics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Eric Marois
- CNRS UPR9022, INSERM U1257, Université de Strasbourg, Strasbourg, France
| | - Catherine Bourgouin
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| |
Collapse
|
42
|
20-Hydroxyecdysone Primes Innate Immune Responses That Limit Bacterial and Malarial Parasite Survival in Anopheles gambiae. mSphere 2020; 5:5/2/e00983-19. [PMID: 32295874 PMCID: PMC7160685 DOI: 10.1128/msphere.00983-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Blood feeding is an integral behavior of mosquitoes to acquire nutritional resources needed for reproduction. This requirement also enables mosquitoes to serve as efficient vectors to acquire and potentially transmit a multitude of mosquito-borne diseases, most notably malaria. Recent studies suggest that mosquito immunity is stimulated following a blood meal, independent of infection status. Since blood feeding promotes production of the hormone 20-hydroxyecdysone (20E), we hypothesized that 20E plays an important role in priming the immune response for pathogen challenge. Here, we examine the immunological effects of priming Anopheles gambiae with 20E prior to pathogen infection, demonstrating a significant reduction in bacteria and Plasmodium berghei survival in the mosquito host. Transcriptome sequencing (RNA-seq) analysis following 20E treatment identifies several known 20E-regulated genes, as well as several immune genes with previously reported function in antipathogen defense. Together, these data demonstrate that 20E influences cellular immune function and antipathogen immunity following mosquito blood feeding, arguing the importance of hormones in the regulation of mosquito innate immune function.IMPORTANCE Blood feeding is required to provide nutrients for mosquito egg production and serves as a mechanism to acquire and transmit pathogens. Shortly after a blood meal is taken, there is a peak in the production of 20-hydroxyecdysone (20E), a mosquito hormone that initiates physiological changes, including yolk protein production and mating refractoriness. Here, we examine additional roles of 20E in the regulation of mosquito immunity, demonstrating that priming the immune system with 20E increases mosquito resistance to pathogens. We identify differentially expressed genes in response to 20E treatment, including several involved in innate immune function as well as lipid metabolism and transport. Together, these data argue that 20E stimulates mosquito cellular immune function and innate immunity shortly after blood feeding.
Collapse
|
43
|
Reduced Function of the Glutathione S-Transferase S1 Suppresses Behavioral Hyperexcitability in Drosophila Expressing Mutant Voltage-Gated Sodium Channels. G3-GENES GENOMES GENETICS 2020; 10:1327-1340. [PMID: 32054635 PMCID: PMC7144092 DOI: 10.1534/g3.119.401025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Voltage-gated sodium (Nav) channels play a central role in the generation and propagation of action potentials in excitable cells such as neurons and muscles. To determine how the phenotypes of Nav-channel mutants are affected by other genes, we performed a forward genetic screen for dominant modifiers of the seizure-prone, gain-of-function Drosophila melanogaster Nav-channel mutant, paraShu. Our analyses using chromosome deficiencies, gene-specific RNA interference, and single-gene mutants revealed that a null allele of glutathione S-transferase S1 (GstS1) dominantly suppresses paraShu phenotypes. Reduced GstS1 function also suppressed phenotypes of other seizure-prone Nav-channel mutants, paraGEFS+ and parabss. Notably, paraShu mutants expressed 50% less GstS1 than wild-type flies, further supporting the notion that paraShu and GstS1 interact functionally. Introduction of a loss-of-function GstS1 mutation into a paraShu background led to up- and down-regulation of various genes, with those encoding cytochrome P450 (CYP) enzymes most significantly over-represented in this group. Because GstS1 is a fly ortholog of mammalian hematopoietic prostaglandin D synthase, and in mammals CYPs are involved in the oxygenation of polyunsaturated fatty acids including prostaglandins, our results raise the intriguing possibility that bioactive lipids play a role in GstS1-mediated suppression of paraShu phenotypes.
Collapse
|
44
|
Jiang D, Du X, Wang C, Zhang S, Wang G. CD109 antigen-like gene is induced by ecdysone signaling and involved in the cellular immunity of Helicoverpa armigera. Biosci Biotechnol Biochem 2020; 84:1183-1190. [PMID: 32141410 DOI: 10.1080/09168451.2020.1737504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Cellular immunity is evolutionarily conserved in invertebrates and vertebrates. In insects, cellular immune response is provided by the hemocytes, and its molecular mechanisms are currently not fully understood. Here, we identified a CD109 antigen-like gene (HaCD109) from Helicoverpa armigera which is highly expressed in the hemocytes of larvae. Stimulation by Escherichia coli and chromatography beads significantly upregulated HaCD109 expression. In vivo HaCD109 silencing significantly increased bacterial load in larval hemolymphs and reduced the hemocyte spread. 20-Hydroxyecdysone (20E) can induce HaCD109 expression through its receptors, EcR and USP. In vivo HaCD109 silencing nearly abolished 20E-induced bacterial clearance and hemocyte spread. These results suggested that HaCD109 plays an important role in cellular immunity, and the 20E-induced cellular immune response in H. armigera requires HaCD109 involvement. Our study contributes to the understanding of regulatory mechanisms for innate immune response and provides new insights into the interaction between innate immunity and steroid hormone signaling.
Collapse
Affiliation(s)
- Di Jiang
- College of Nursing, Weifang Medical University, Weifang, China
| | - Xuemeng Du
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Cuiyan Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Shisong Zhang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Gang Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| |
Collapse
|
45
|
Belmonte RL, Corbally MK, Duneau DF, Regan JC. Sexual Dimorphisms in Innate Immunity and Responses to Infection in Drosophila melanogaster. Front Immunol 2020; 10:3075. [PMID: 32076419 PMCID: PMC7006818 DOI: 10.3389/fimmu.2019.03075] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022] Open
Abstract
The sexes show profound differences in responses to infection and the development of autoimmunity. Dimorphisms in immune responses are ubiquitous across taxa, from arthropods to vertebrates. Drosophila melanogaster shows strong sex dimorphisms in immune system responses at baseline, upon pathogenic challenge, and over aging. We have performed an exhaustive survey of peer-reviewed literature on Drosophila immunity, and present a database of publications indicating the sex(es) analyzed in each study. While we found a growing interest in the community in adult immunity and in reporting both sexes, the main body of work in this field uses only one sex, or does not stratify by sex. We synthesize evidence for sexually dimorphic responses to bacterial, viral, and fungal infections. Dimorphisms may be mediated by distinct immune compartments, and we review work on sex differences in behavioral, epithelial, cellular, and systemic (fat body-mediated) immunity. Emerging work on sexually dimorphic aging of immune tissues, immune senescence, and inflammation are examined. We consider evolutionary drivers for sex differences in immune investment, highlight the features of Drosophila biology that make it particularly amenable to studies of immune dimorphisms, and discuss areas for future exploration.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary-Kate Corbally
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David F. Duneau
- Laboratoire Evolution & Diversite Biologique, UMR5174 EDB, CNRS, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Jennifer C. Regan
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
46
|
A mosquito juvenile hormone binding protein (mJHBP) regulates the activation of innate immune defenses and hemocyte development. PLoS Pathog 2020; 16:e1008288. [PMID: 31961911 PMCID: PMC6994123 DOI: 10.1371/journal.ppat.1008288] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 01/31/2020] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
Insects rely on the innate immune system for defense against pathogens, some aspects of which are under hormonal control. Here we provide direct experimental evidence showing that the juvenile hormone-binding protein (mJHBP) of Aedes aegypti is required for the regulation of innate immune responses and the development of mosquito blood cells (hemocytes). Using an mJHBP-deficient mosquito line generated by means of CRISPR-Cas9 gene editing technology we uncovered a mutant phenotype characterized by immunosuppression at the humoral and cellular levels, which profoundly affected susceptibility to bacterial infection. Bacteria-challenged mosquitoes exhibited significantly higher levels of septicemia and mortality relative to the wild type (WT) strain, delayed expression of antimicrobial peptides (AMPs), severe developmental dysregulation of embryonic and larval hemocytes (reduction in the total number of hemocytes) and increased differentiation of the granulocyte lineage. Interestingly, injection of recombinant wild type mJHBP protein into adult females three-days before infection was sufficient to restore normal immune function. Similarly, injection of mJHBP into fourth-instar larvae fully restored normal larval/pupal hemocyte populations in emerging adults. More importantly, the recovery of normal immuno-activation and hemocyte development requires the capability of mJHBP to bind JH III. These results strongly suggest that JH III functions in mosquito immunity and hemocyte development in a manner that is perhaps independent of canonical JH signaling, given the lack of developmental and reproductive abnormalities. Because of the prominent role of hemocytes as regulators of mosquito immunity, this novel discovery may have broader implications for the understanding of vector endocrinology, hemocyte development, vector competence and disease transmission. There are many unanswered questions concerning the nature of immune responses of mosquitoes to bacteria, viruses and parasites. This is important because a variety of human pathogens are transmitted by mosquitoes during the process of consuming blood. Much of mosquito physiology is under the control of hormones and we aim to understand a potential role for an important hormone known as juvenile hormone in anti-bacterial immunity. We have produced a strain of the mosquito, Aedes aegypti, that is deficient in the production of a protein that circulates in the blood while carrying juvenile hormone. This strain is shown to have less ability to control bacterial infection, to have lower levels of proteins involved in immunity and to have smaller numbers of blood cells that are known to be important in the mosquito immune response. If the protein is administered to the deficient strain by injection, the immune response and blood cell numbers return to near-normal levels. Other results suggest that the association of the protein with juvenile hormone is important for its ability to function in the immune system. Overall, this study describes an important new protein regulator of mosquito immunity and a potential role of juvenile hormone in this process.
Collapse
|
47
|
Simu-dependent clearance of dying cells regulates macrophage function and inflammation resolution. PLoS Biol 2019; 17:e2006741. [PMID: 31086359 PMCID: PMC6516643 DOI: 10.1371/journal.pbio.2006741] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 04/10/2019] [Indexed: 12/30/2022] Open
Abstract
Macrophages encounter and clear apoptotic cells during normal development and homeostasis, including at numerous sites of pathology. Clearance of apoptotic cells has been intensively studied, but the effects of macrophage-apoptotic cell interactions on macrophage behaviour are poorly understood. Using Drosophila embryos, we have exploited the ease of manipulating cell death and apoptotic cell clearance in this model to identify that the loss of the apoptotic cell clearance receptor Six-microns-under (Simu) leads to perturbation of macrophage migration and inflammatory responses via pathological levels of apoptotic cells. Removal of apoptosis ameliorates these phenotypes, while acute induction of apoptosis phenocopies these defects and reveals that phagocytosis of apoptotic cells is not necessary for their anti-inflammatory action. Furthermore, Simu is necessary for clearance of necrotic debris and retention of macrophages at wounds. Thus, Simu is a general detector of damaged self and represents a novel molecular player regulating macrophages during resolution of inflammation.
Collapse
|
48
|
Ma H, Abbas MN, Zhang K, Hu X, Xu M, Liang H, Kausar S, Yang L, Cui H. 20-Hydroxyecdysone regulates the transcription of the lysozyme via Broad-Complex Z2 gene in silkworm, Bombyx mori. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 94:66-72. [PMID: 30716346 DOI: 10.1016/j.dci.2019.01.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 06/09/2023]
Abstract
Broad-Complex Z2 (Br-C Z2) is an ecdysone inducible transcription factor that regulates physiological, innate immune and developmental events in insects. Here, we identified an orthologue of Br-C Z2 from silkworm, Bombyx mori (BmBr-C Z2) to study its involvement in immune responses. The quantitative real-time PCR analysis revealed that BmBr-C Z2 was expressed ubiquitously in all tested tissues under normal physiological conditions. Further, developmental profile displayed that BmBr-C Z2 expression was detectable in different developmental stages, however the gene's expression was highest in the molting and pre-pupal stages. Administration of 20-hydroxyecdysone (20E) enhanced the expression levels of BmBr-C Z2 in hemocytes. The challenge with pathogens and pathogen associated molecular patterns (PAMPs) also upregulated the mRNA levels of BmBr-C Z2 in hemocytes when compared with the control. By contrast, the ectopic expression of BmBr-C Z2 remarkably increased the production of antimicrobial peptides, while the knock-down of this gene by double stranded RNA decreased their production. Dual-luciferase assay exhibited that BmBr-C Z2 induced the expression of lysozyme by directly binding to its promoter region. The treatment of Escherichia coli following the knock-down of BmBr-C Z2 strongly reduced the survival rate of silkworm larvae. These results suggest that BmBr-C Z2 plays an important biological role in the innate immune responses of silkworm by regulating immune-related genes.
Collapse
Affiliation(s)
- Hanxiu Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Xiaosong Hu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Man Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Hanghua Liang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China; Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, 400716, Chongqing, China; Southwest University Engineering Research Center for Cancer Biomedical and Translational Medicine, 400716, Chongqing, China
| |
Collapse
|
49
|
Troha K, Buchon N. Methods for the study of innate immunity in Drosophila melanogaster. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e344. [PMID: 30993906 DOI: 10.1002/wdev.344] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
From flies to humans, many components of the innate immune system have been conserved during metazoan evolution. This foundational observation has allowed us to develop Drosophila melanogaster, the fruit fly, into a powerful model to study innate immunity in animals. Thanks to an ever-growing arsenal of genetic tools, an easily manipulated genome, and its winning disposition, Drosophila is now employed to study not only basic molecular mechanisms of pathogen recognition and immune signaling, but also the nature of physiological responses activated in the host by microbial challenge and how dysregulation of these processes contributes to disease. Here, we present a collection of methods and protocols to challenge the fly with an assortment of microbes, both systemically and orally, and assess its humoral, cellular, and epithelial response to infection. Our review covers techniques for measuring the reaction to microbial infection both qualitatively and quantitatively. Specifically, we describe survival, bacterial load, BLUD (a measure of disease tolerance), phagocytosis, melanization, clotting, and ROS production assays, as well as efficient protocols to collect hemolymph and measure immune gene expression. We also offer an updated catalog of online resources and a collection of popular reporter lines and mutants to facilitate research efforts. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.
Collapse
Affiliation(s)
- Katia Troha
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York
| | - Nicolas Buchon
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York
| |
Collapse
|
50
|
Banerjee U, Girard JR, Goins LM, Spratford CM. Drosophila as a Genetic Model for Hematopoiesis. Genetics 2019; 211:367-417. [PMID: 30733377 PMCID: PMC6366919 DOI: 10.1534/genetics.118.300223] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
In this FlyBook chapter, we present a survey of the current literature on the development of the hematopoietic system in Drosophila The Drosophila blood system consists entirely of cells that function in innate immunity, tissue integrity, wound healing, and various forms of stress response, and are therefore functionally similar to myeloid cells in mammals. The primary cell types are specialized for phagocytic, melanization, and encapsulation functions. As in mammalian systems, multiple sites of hematopoiesis are evident in Drosophila and the mechanisms involved in this process employ many of the same molecular strategies that exemplify blood development in humans. Drosophila blood progenitors respond to internal and external stress by coopting developmental pathways that involve both local and systemic signals. An important goal of these Drosophila studies is to develop the tools and mechanisms critical to further our understanding of human hematopoiesis during homeostasis and dysfunction.
Collapse
Affiliation(s)
- Utpal Banerjee
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
- Molecular Biology Institute, University of California, Los Angeles, California 90095
- Department of Biological Chemistry, University of California, Los Angeles, California 90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Juliet R Girard
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Lauren M Goins
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| | - Carrie M Spratford
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California 90095
| |
Collapse
|