1
|
Yao X, Lin L, Ye Z, Huo M, Jin P, Ma F. NF-κB/Relish readjusts miR-100 expression and recovers immune homeostasis in Drosophila melanogaster. INSECT SCIENCE 2024. [PMID: 39688880 DOI: 10.1111/1744-7917.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
The regulation and maintenance of immune homeostasis are essential for animal survival, but the molecular mechanisms are not fully understood. Here, we used the model organism Drosophila melanogaster to uncover a potential mechanism by which the nuclear factor-κB transcription factor Relish and miR-100 cooperatively regulate innate immune homeostasis. We first demonstrated in vitro and in vivo that miR-100 can negatively regulate the immune responses of the Imd pathway by inhibiting the expression of TAK1-associated binding protein 2 (Tab2) gene. Second, we found that Relish, an important transcription factor in the Drosophila Imd pathway, could not only modulate the expressions of antimicrobial peptides (AMPs) to promote immune responses, but also bind to the promoter region of miR-100 and activate its transcription to inhibit immune responses. Third, the dynamic expression of genes profiling indicated that the Relish/miR-100/Tab2 regulatory axis could contribute to innate immune homeostasis in Drosophila. Together, our findings reveal the dual role of Relish in immune regulation, that is, Relish promotes the expression of AMPs to resist pathogen infection in the early immune response, while in the late immune stages, Relish readjusts the expression of miR-100 to negatively control immune responses to avoid excessive immunity thus maintaining immunohomeostasis. Meanwhile, our study provides a new perspective for further understanding the complex regulatory mechanism of immune homeostasis in animals.
Collapse
Affiliation(s)
- Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Zifeng Ye
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Miaomiao Huo
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| |
Collapse
|
2
|
Liu S, Raheel Tariq M, Zhang Q, Wang H, Wang F, Zheng C, Li K, Zhuang Z, Wang L. Dietary Influence on Growth, Physicochemical Stability, and Antimicrobial Mechanisms of Antimicrobial Peptides in Black Soldier Fly Larvae. INSECTS 2024; 15:872. [PMID: 39590471 PMCID: PMC11595210 DOI: 10.3390/insects15110872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024]
Abstract
Safe antibiotic substitutes are needed given the rise in antimicrobial resistance, environmental contamination, and stringent antibiotic regulations. Insect-derived antimicrobial peptides (AMPs) are promising candidates due to their antimicrobial activity, stability, and safety. This study investigates the antimicrobial mechanism of crude AMP extracts and their physicochemical characteristics in black soldier fly larvae (BSFL). The results indicated that BSFL reared on a wheat bran diet exhibited significantly improved growth performance and AMP production when compared to the other three diets. AMP extracts showed enhanced antimicrobial activity and physicochemical stability, including temperatures and metal ions except Cu+. Moreover, AMP extracts disrupted the cell membrane and inhibited the cell cycle of Staphylococcus aureus (S. aureus), thus exhibiting antimicrobial activity. Furthermore, transcriptomic and KEGG enrichment analyses identified 509 differentially expressed genes (DEGs) related to the Toll and IMD signaling pathways. STRING and GeneMANIA analyses confirmed the association of these pathways with immune response and AMP secretion. qRT-PCR results showed elevated expression of immune genes (GNBP3, NFKBIA, GADD45, and Spz) in BSFL following S. aureus immunization, consistent with RNA-seq findings. These findings offer a valuable reference for using AMPs as antibiotic substitutes in animal feeds and highlight the need for further research on AMP purification and the synergistic regulation of protein synthesis and AMP production in BSFL.
Collapse
Affiliation(s)
- Shaojuan Liu
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| | - Muhammad Raheel Tariq
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 524088, China
| | - Qihui Zhang
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 524088, China
| | - Hui Wang
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| | - Fei Wang
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| | - Chaozhong Zheng
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| | - Kuntai Li
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
- College of Biological Sciences and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zhikai Zhuang
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| | - Leiyu Wang
- Agricultural Products Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China; (S.L.); (M.R.T.); (C.Z.)
| |
Collapse
|
3
|
Asgari D, Tate AT. How the Structure of Signaling Regulation Evolves: Insights from an Evolutionary Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619883. [PMID: 39484560 PMCID: PMC11526956 DOI: 10.1101/2024.10.23.619883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
To remain responsive to environmental changes, signaling pathways attenuate their activity with negative feedback loops (NFLs), where proteins produced upon stimulation downregulate the response. NFLs function both upstream of signaling to reduce input and downstream to reduce output. Unlike upstream NFLs, downstream NFLs directly regulate gene expression without the involvement of intermediate proteins. Thus, we hypothesized that downstream NFLs evolve under more stringent selection than upstream NFLs. Indeed, genes encoding downstream NFLs exhibit a slower evolutionary rate than upstream genes. Such differences in selective pressures could result in the robust evolution of downstream NFLs while making the evolution of upstream NFLs more sensitive to changes in signaling proteins and stimuli. Here, we test these assumptions within the context of immune signaling. Our minimal model of immune signaling predicts robust evolution of downstream NFLs to changes in model parameters. This is consistent with their critical role in regulating signaling and the conservative rate of evolution. Furthermore, we show that the number of signaling steps needed to activate a downstream NFL is influenced by the cost of signaling. Our model predicts that upstream NFLs are more likely to evolve under a shorter half-life of signaling proteins, absence of host-pathogen co-evolution, and a high infection rate. Although it has been proposed that NFLs evolve to reduce the cost of signaling, we show that a high cost does not necessarily predict the evolution of upstream NFLs. The insights from our model have broad implications for understanding the evolution of regulatory mechanisms across signaling pathways.
Collapse
Affiliation(s)
- Danial Asgari
- Department of Biological Sciences, Vanderbilt University, Nashville TN 37232
- Evolutionary Studies Initiative, Vanderbilt University, Nashville TN 37232
| | - Ann T. Tate
- Department of Biological Sciences, Vanderbilt University, Nashville TN 37232
- Evolutionary Studies Initiative, Vanderbilt University, Nashville TN 37232
| |
Collapse
|
4
|
Asgari D, Stewart AJ, Meisel RP. The role of uncertainty and negative feedback loops in the evolution of induced immune defenses. G3 (BETHESDA, MD.) 2024; 14:jkae182. [PMID: 39106431 PMCID: PMC11457078 DOI: 10.1093/g3journal/jkae182] [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: 06/13/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Organisms use constitutive or induced defenses against pathogens and other external threats. Constitutive defenses are constantly on, whereas induced defenses are activated when needed. Each of these strategies has costs and benefits, which can affect the type of defense that evolves in response to pathogens. In addition, induced defenses are usually regulated by multiple negative feedback mechanisms that prevent overactivation of the immune response. However, it is unclear how negative feedback affects the costs, benefits, and evolution of induced responses. To address this gap, we developed a mechanistic model of the well-characterized Drosophila melanogaster immune signaling network that includes 3 separate mechanisms of negative feedback as a representative of the widespread phenomenon of multilevel regulation of induced responses. We show that, under stochastic fly-bacteria encounters, an induced defense is favored when bacterial encounters are rare or uncertain, but in ways that depend on the bacterial proliferation rate. Our model also predicts that the specific negative regulators that optimize the induced response depend on the bacterial proliferation rate, linking negative feedback mechanisms to the factors that favor induction.
Collapse
Affiliation(s)
- Danial Asgari
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Alexander J Stewart
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9AJ, UK
| | - Richard P Meisel
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
5
|
Aalto AL, Luukkonen V, Meinander A. Ubiquitin signalling in Drosophila innate immune responses. FEBS J 2024; 291:4397-4413. [PMID: 38069549 DOI: 10.1111/febs.17028] [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: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Cells respond to invading pathogens and danger signals from the environment by adapting gene expression to meet the need for protective effector molecules. While this innate immune response is required for the cell and the organism to recover, excess immune activation may lead to loss of homeostasis, thereby promoting chronic inflammation and cancer progression. The molecular basis of innate immune defence is comprised of factors promoting survival and proliferation, such as cytokines, antimicrobial peptides and anti-apoptotic proteins. As the molecular mechanisms regulating innate immune responses are conserved through evolution, the fruit fly Drosophila melanogaster serves as a convenient, affordable and ethical model organism to enhance understanding of immune signalling. Fly immunity against bacterial infection is built up by both cellular and humoral responses, where the latter is regulated by the Imd and Toll pathways activating NF-κB transcription factors Relish, Dorsal and Dif, as well as JNK activation and JAK/STAT signalling. As in mammals, the Drosophila innate immune signalling pathways are characterised by ubiquitination of signalling molecules followed by ubiquitin receptors binding to the ubiquitin chains, as well as by rapid changes in protein levels by ubiquitin-mediated targeted proteasomal and lysosomal degradation. In this review, we summarise the molecular signalling pathways regulating immune responses to pathogen infection in Drosophila, with a focus on ubiquitin-dependent control of innate immunity and inflammatory signalling.
Collapse
Affiliation(s)
- Anna L Aalto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| | - Veera Luukkonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | - Annika Meinander
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship, Åbo Akademi University, Turku, Finland
| |
Collapse
|
6
|
Hossain Mollah J, Hatimuria A, Kumar Chauhan V. Transcriptomic analysis of Bombyx mori in its early larval stage (2 nd instar) of development upon Nosema bombycis transovarial infection. J Invertebr Pathol 2024; 206:108157. [PMID: 38908473 DOI: 10.1016/j.jip.2024.108157] [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: 01/01/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
The infection caused by Nosema bombycis often known as pebrine, is a devastating sericulture disease. The infection can be transmitted to the next generation through eggs laid by infected female Bombyx mori moths (transovarial) as well as with N. bombycis contaminated food (horizontal). Most diagnoses were carried out in the advanced stages of infection until the time that infection might spread to other healthy insects. Hence, early diagnosis of pebrine is of utmost importance to quarantine infected larvae from uninfected silkworm batches and stop further spread of the infection. The findings of our study provide an insight into how the silkworm larval host defence system was activated against early N. bombycis transovarial infection. The results obtained from transcriptome analysis of infected 2nd instar larvae revealed significant (adjusted P-value < 0.05) expression of 1888 genes of which 801 genes were found to be upregulated and 1087 genes were downregulated when compared with the control. Pathway analysis indicated activation of the immune deficiency (IMD) pathway, which shows a potential immune defence response against pebrine infection as well as suppression of the melanin synthesis pathway due to lower expression of prophenoloxidase activating enzyme (PPAE). Liquid chromatography mass spectrometry (LC-MS/MS) analysis of haemolymph from infected larvae shows the secretion of serpin binding protein of N. bombycis which might be involved in the suppression of the melanization pathway. Moreover, among the differentially expressed genes, we found that LPMC-61, yellow-y, gasp and osiris 9 can be utilised as potential markers for early diagnosis of transovarial pebrine infection in B. mori. Physiological as well as biochemical roles and functions of many of the essential genes are yet to be established, and enlightened research will be required to characterize the products of these genes.
Collapse
Affiliation(s)
- Jahid Hossain Mollah
- Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati, Santiniketan, West Bengal-731235, India
| | - Arindam Hatimuria
- Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati, Santiniketan, West Bengal-731235, India
| | - Vinod Kumar Chauhan
- Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati, Santiniketan, West Bengal-731235, India.
| |
Collapse
|
7
|
Fioriti F, Rifflet A, Gomperts Boneca I, Zugasti O, Royet J. Bacterial peptidoglycan serves as a critical modulator of the gut-immune-brain axis in Drosophila. Brain Behav Immun 2024; 119:878-897. [PMID: 38710338 DOI: 10.1016/j.bbi.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/26/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
Metabolites and compounds derived from gut-associated bacteria can modulate numerous physiological processes in the host, including immunity and behavior. Using a model of oral bacterial infection, we previously demonstrated that gut-derived peptidoglycan (PGN), an essential constituent of the bacterial cell envelope, influences female fruit fly egg-laying behavior by activating the NF-κB cascade in a subset of brain neurons. These findings underscore PGN as a potential mediator of communication between gut bacteria and the brain in Drosophila, prompting further investigation into its impact on all brain cells. Through high-resolution mass spectrometry, we now show that PGN fragments produced by gut bacteria can rapidly reach the central nervous system. In Addition, by employing a combination of whole-genome transcriptome analyses, comprehensive genetic assays, and reporter gene systems, we reveal that gut bacterial infection triggers a PGN dose-dependent NF-κB immune response in perineurial glia, forming the continuous outer cell layer of the blood-brain barrier. Furthermore, we demonstrate that persistent PGN-dependent NF-κB activation in perineurial glial cells correlates with a reduction in lifespan and early neurological decline. Overall, our findings establish gut-derived PGN as a critical mediator of the gut-immune-brain axis in Drosophila.
Collapse
Affiliation(s)
- Florent Fioriti
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France
| | - Aline Rifflet
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, 75015 Paris, France
| | - Ivo Gomperts Boneca
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, INSERM U1306, 75015 Paris, France
| | - Olivier Zugasti
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France.
| | - Julien Royet
- Institut de Biologie du Développement de Marseille, Aix-Marseille Université, CNRS UMR 7288 Marseille, France.
| |
Collapse
|
8
|
Hajra D, Kirthivasan N, Chakravortty D. Symbiotic Synergy from Sponges to Humans: Microflora-Host Harmony Is Crucial for Ensuring Survival and Shielding against Invading Pathogens. ACS Infect Dis 2024; 10:317-336. [PMID: 38170903 DOI: 10.1021/acsinfecdis.3c00554] [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] [Indexed: 01/05/2024]
Abstract
Gut microbiota plays several roles in the host organism's metabolism and physiology. This phenomenon holds across different species from different kingdoms and classes. Different species across various classes engage in continuous crosstalk via various mechanisms with their gut microbiota, ensuring homeostasis of the host. In this Review, the diversity of the microflora, the development of the microflora in the host, its regulations by the host, and its functional implications on the host, especially in the context of dysbiosis, are discussed across different organisms from sponges to humans. Overall, our review aims to address the indispensable nature of the microbiome in the host's survival, fitness, and protection against invading pathogens.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Nikhita Kirthivasan
- Undergraduate Programme, Indian Institute of Science, Bangalore, Karnataka-560012, India
| | - Dipshikha Chakravortty
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore, Karnataka-560012, India
| |
Collapse
|
9
|
Liu M, Yang S, Yang J, Feng P, Luo F, Zhang Q, Yang L, Jiang H. BubR1 controls starvation-induced lipolysis via IMD signaling pathway in Drosophila. Aging (Albany NY) 2024; 16:3257-3279. [PMID: 38334966 PMCID: PMC10929803 DOI: 10.18632/aging.205533] [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: 07/14/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Lipolysis, the key process releasing fat acids to generate energy in adipose tissues, correlates with starvation resistance. Nevertheless, its detail mechanisms remain elusive. BubR1, an essential mitotic regulator, ensures proper chromosome alignment and segregation during mitosis, but its physiological functions are largely unknown. Here, we use Drosophila adult fat body, the major lipid storage organ, to study the functions of BubR1 in lipolysis. We show that both whole body- and fat body-specific BubR1 depletions increase lipid degradation and shorten the lifespan under fasting but not feeding. Relish, the conserved regulator of IMD signaling pathway, acts as the downstream target of BubR1 to control the expression level of Bmm and modulate the lipolysis upon fasting. Thus, our study reveals new functions of BubR1 in starvation-induced lipolysis and provides new insights into the molecular mechanisms of lipolysis mediated by IMD signaling pathway.
Collapse
Affiliation(s)
- Mengyou Liu
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shengye Yang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingsi Yang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ping Feng
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feng Luo
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiaoqiao Zhang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li Yang
- Department of Gastroenterology and Hepatology and Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hao Jiang
- Laboratory for Aging and Cancer Research, Frontiers Science Center Disease-related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Critchlow JT, Prakash A, Zhong KY, Tate AT. Mapping the functional form of the trade-off between infection resistance and reproductive fitness under dysregulated immune signaling. PLoS Pathog 2024; 20:e1012049. [PMID: 38408106 PMCID: PMC10919860 DOI: 10.1371/journal.ppat.1012049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/07/2024] [Accepted: 02/15/2024] [Indexed: 02/28/2024] Open
Abstract
Immune responses benefit organismal fitness by clearing parasites but also exact costs associated with immunopathology and energetic investment. Hosts manage these costs by tightly regulating the induction of immune signaling to curtail excessive responses and restore homeostasis. Despite the theoretical importance of turning off the immune response to mitigate these costs, experimentally connecting variation in the negative regulation of immune responses to organismal fitness remains a frontier in evolutionary immunology. In this study, we used a dose-response approach to manipulate the RNAi-mediated knockdown efficiency of cactus (IκBα), a central regulator of Toll pathway signal transduction in flour beetles (Tribolium castaneum). By titrating cactus activity across four distinct levels, we derived the shape of the relationship between immune response investment and traits associated with host fitness, including infection susceptibility, lifespan, fecundity, body mass, and gut homeostasis. Cactus knock-down increased the overall magnitude of inducible immune responses and delayed their resolution in a dsRNA dose-dependent manner, promoting survival and resistance following bacterial infection. However, these benefits were counterbalanced by dsRNA dose-dependent costs to lifespan, fecundity, body mass, and gut integrity. Our results allowed us to move beyond the qualitative identification of a trade-off between immune investment and fitness to actually derive its functional form. This approach paves the way to quantitatively compare the evolution and impact of distinct regulatory elements on life-history trade-offs and fitness, filling a crucial gap in our conceptual and theoretical models of immune signaling network evolution and the maintenance of natural variation in immune systems.
Collapse
Affiliation(s)
- Justin T Critchlow
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Arun Prakash
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Katherine Y Zhong
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ann T Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Evolutionary Studies Institute, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
11
|
Pan W, Yao X, Lin L, Liu X, Jin P, Ma F. The Relish/miR-275/Dredd mediated negative feedback loop is crucial to restoring immune homeostasis of Drosophila Imd pathway. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 162:104013. [PMID: 37804878 DOI: 10.1016/j.ibmb.2023.104013] [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: 05/15/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
The NF-κB/Relish, as a core transcription factor of Drosophila immune deficiency (Imd) pathway, activates the transcriptions of antimicrobial peptides (AMPs) to combat gram-negative bacterial infections, but its role in regulating miRNA expression during immune response has less been reported. We here describe a negative feedback loop of Imd signaling mediated by Relish/miR-275/Dredd that controls Drosophila immune homeostasis after Escherichia coli (E. coli) infection. Our results demonstrate that Relish may directly activate the transcription of miR-275 via binding to its promoter in vitro and vivo, particularly miR-275 further inhibits the expression of Dredd through binding to its 3'UTR to negatively control Drosophila Imd immune response. Remarkably, the ectopic expression of miR-275 significantly reduces Drosophila lifespan. More importantly, our work uncovers a new mechanism by which Relish can flexibly switch its role to maintain Drosophila immune response and homeostasis during infection. Collectively, our study not only reveals the functional duality of Relish in regulating immune response of Drosophila Imd pathway, but also provides a new insight into the maintenance of animal innate immune homeostasis.
Collapse
Affiliation(s)
- Wanwan Pan
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xiaoqi Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
12
|
Yao X, Ni J, Lin L, Jin P, Ma F. The NF-κB/Relish Activates miR-308 to Negatively Regulate Imd Pathway Immune Signaling in Drosophila. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:591-600. [PMID: 37358278 DOI: 10.4049/jimmunol.2200680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 06/02/2023] [Indexed: 06/27/2023]
Abstract
The strength and duration of the NF-κB signaling response must be tightly modulated to avoid an inadequate or excessive immune response. Relish, a core NF-κB transcription factor of the Drosophila Imd pathway, can control the expression of antimicrobial peptides, including Dpt and AttA, to defend against Gram-negative bacterial infections, but whether Relish may regulate miRNA expression to participate in the immune response remains unclear. In this study, taking advantage of Drosophila S2 cells and different overexpression/knockout/knockdown flies, we first found that Relish could directly activate the expression of miR-308 to negatively regulate the immune response and promote the survival of Drosophila during Enterobacter cloacae infection. Second, our results demonstrated that Relish-mediated expression of miR-308 could suppress target gene Tab2 to attenuate the Drosophila Imd pathway signal during the middle and late stages of the immune response. Third, we detected the dynamic expression patterns of Dpt, AttA, Relish, miR-308, and Tab2 in wild-type flies after E. coli infection, which further revealed that the feedback regulatory loop of Relish-miR-308-Tab2 plays a crucial role in the immune response and homeostasis maintenance of the Drosophila Imd pathway. Overall, our present study not only illustrates an important mechanism by which this Relish-miR-308-Tab2 regulatory axis can negatively control the Drosophila immune response and participate in homeostasis maintenance but also provides new insights into the dynamic regulation of the NF-κB/miRNA expression network of animal innate immunity.
Collapse
Affiliation(s)
- Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Jiajia Ni
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| |
Collapse
|
13
|
Critchlow JT, Prakash A, Zhong KY, Tate AT. Mapping the functional form of the trade-off between infection resistance and reproductive fitness under dysregulated immune signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552815. [PMID: 37645726 PMCID: PMC10461925 DOI: 10.1101/2023.08.10.552815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Immune responses benefit organismal fitness by clearing parasites but also exact costs associated with immunopathology and energetic investment. Hosts manage these costs by tightly regulating the induction of immune signaling to curtail excessive responses and restore homeostasis. Despite the theoretical importance of turning off the immune response to mitigate these costs, experimentally connecting variation in the negative regulation of immune responses to organismal fitness remains a frontier in evolutionary immunology. In this study, we used a dose-response approach to manipulate the RNAi-mediated knockdown efficiency of cactus (IκBα), a central regulator of Toll pathway signal transduction in flour beetles (Tribolium castaneum). By titrating cactus activity along a continuous gradient, we derived the shape of the relationship between immune response investment and traits associated with host fitness, including infection susceptibility, lifespan, fecundity, body mass, and gut homeostasis. Cactus knock-down increased the overall magintude of inducible immune responses and delayed their resolution in a dsRNA dose-dependent manner, promoting survival and resistance following bacterial infection. However, these benefits were counterbalanced by dsRNA dose-dependent costs to lifespan, fecundity, body mass, and gut integrity. Our results allowed us to move beyond the qualitative identification of a trade-off between immune investment and fitness to actually derive its functional form. This approach paves the way to quantitatively compare the evolution and impact of distinct regulatory elements on life-history trade-offs and fitness, filling a crucial gap in our conceptual and theoretical models of immune signaling network evolution and the maintenance of natural variation in immune systems.
Collapse
Affiliation(s)
- Justin T. Critchlow
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Arun Prakash
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Katherine Y. Zhong
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ann T. Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Evolutionary Studies Institute, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
14
|
Bossen J, Kühle JP, Roeder T. The tracheal immune system of insects - A blueprint for understanding epithelial immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103960. [PMID: 37235953 DOI: 10.1016/j.ibmb.2023.103960] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
The unique design of respiratory organs in multicellular organisms makes them prone to infection by pathogens. To cope with this vulnerability, highly effective local immune systems evolved that are also operative in the tracheal system of insects. Many pathogens and parasites (including viruses, bacteria, fungi, and metazoan parasites) colonize the trachea or invade the host via this route. Currently, only two modules of the tracheal immune system have been characterized in depth: 1) Immune deficiency pathway-mediated activation of antimicrobial peptide gene expression and 2) local melanization processes that protect the structure from wounding. There is an urgent need to increase our understanding of the architecture of tracheal immune systems, especially regarding those mechanisms that enable the maintenance of immune homeostasis. This need for new studies is particularly exigent for species other than Drosophila.
Collapse
Affiliation(s)
- Judith Bossen
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany
| | - Jan-Philip Kühle
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany
| | - Thomas Roeder
- Kiel University, Zoology, Dept, Molecular Physiology, Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Germany.
| |
Collapse
|
15
|
Tan FHP, Azzam G, Najimudin N, Shamsuddin S, Zainuddin A. Behavioural Effects and RNA-seq Analysis of Aβ42-Mediated Toxicity in a Drosophila Alzheimer's Disease Model. Mol Neurobiol 2023:10.1007/s12035-023-03368-x. [PMID: 37145377 DOI: 10.1007/s12035-023-03368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/22/2023] [Indexed: 05/06/2023]
Abstract
Alzheimer's disease (AD) is the most common neurological ailment worldwide. Its process comprises the unique aggregation of extracellular senile plaques composed of amyloid-beta (Aβ) in the brain. Aβ42 is the most neurotoxic and aggressive of the Aβ42 isomers released in the brain. Despite much research on AD, the complete pathophysiology of this disease remains unknown. Technical and ethical constraints place limits on experiments utilizing human subjects. Thus, animal models were used to replicate human diseases. The Drosophila melanogaster is an excellent model for studying both physiological and behavioural aspects of human neurodegenerative illnesses. Here, the negative effects of Aβ42-expression on a Drosophila AD model were investigated through three behavioural assays followed by RNA-seq. The RNA-seq data was verified using qPCR. AD Drosophila expressing human Aβ42 exhibited degenerated eye structures, shortened lifespan, and declined mobility function compared to the wild-type Control. RNA-seq revealed 1496 genes that were differentially expressed from the Aβ42-expressing samples against the control. Among the pathways that were identified from the differentially expressed genes include carbon metabolism, oxidative phosphorylation, antimicrobial peptides, and longevity-regulating pathways. While AD is a complicated neurological condition whose aetiology is influenced by a number of factors, it is hoped that the current data will be sufficient to give a general picture of how Aβ42 influences the disease pathology. The discovery of molecular connections from the current Drosophila AD model offers fresh perspectives on the usage of this Drosophila which could aid in the discovery of new anti-AD medications.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia.
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Ghows Azzam
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia.
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia.
- Malaysia Genome and Vaccine Institute (MGVI), National Institutes of Biotechnology Malaysia (NIBM), Jalan Bangi, 43000, Kajang, Selangor, Malaysia.
| | - Nazalan Najimudin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- USM-RIKEN Interdisciplinary Centre for Advanced Sciences (URICAS), Universiti Sains Malaysia, 11800, Penang, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800, Penang, Malaysia
| | - Azalina Zainuddin
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Gu J, Zhang P, Yao Z, Li X, Zhang H. BdNub Is Essential for Maintaining gut Immunity and Microbiome Homeostasis in Bactrocera dorsalis. INSECTS 2023; 14:178. [PMID: 36835747 PMCID: PMC9964267 DOI: 10.3390/insects14020178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Insects face immune challenges posed by invading and indigenous bacteria. They rely on the immune system to clear these microorganisms. However, the immune response can be harmful to the host. Therefore, fine-tuning the immune response to maintain tissue homeostasis is of great importance to the survival of insects. The Nub gene of the OCT/POU family regulates the intestinal IMD pathway. However, the role of the Nub gene in regulating host microbiota remains unstudied. Here, a combination of bioinformatic tools, RNA interference, and qPCR methods were adopted to study BdNub gene function in Bactrocera dorsalis gut immune system. It's found that BdNubX1, BdNubX2, and antimicrobial peptides (AMPs), including Diptcin (Dpt), Cecropin (Cec), AttcinA (Att A), AttcinB (Att B) and AttcinC (Att C) are significantly up-regulated in Tephritidae fruit fly Bactrocera dorsalis after gut infection. Silencing BdNubX1 leads to down-regulated AMPs expression, while BdNubX2 RNAi leads to increased expression of AMPs. These results indicate that BdNubX1 is a positive regulatory gene of the IMD pathway, while BdNubX2 negatively regulates IMD pathway activity. Further studies also revealed that BdNubX1 and BdNubX2 are associated with gut microbiota composition, possibly through regulation of IMD pathway activity. Our results prove that the Nub gene is evolutionarily conserved and participates in maintaining gut microbiota homeostasis.
Collapse
|
17
|
Zeng T, Jaffar S, Xu Y, Qi Y. The Intestinal Immune Defense System in Insects. Int J Mol Sci 2022; 23:ijms232315132. [PMID: 36499457 PMCID: PMC9740067 DOI: 10.3390/ijms232315132] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Over a long period of evolution, insects have developed unique intestinal defenses against invasion by foreign microorganisms, including physical defenses and immune responses. The physical defenses of the insect gut consist mainly of the peritrophic matrix (PM) and mucus layer, which are the first barriers to pathogens. Gut microbes also prevent the colonization of pathogens. Importantly, the immune-deficiency (Imd) pathways produce antimicrobial peptides to eliminate pathogens; mechanisms related to reactive oxygen species are another important pathway for insect intestinal immunity. The janus kinase/STAT signaling pathway is involved in intestinal immunity by producing bactericidal substances and regulating tissue repair. Melanization can produce many bactericidal active substances into the intestine; meanwhile, there are multiple responses in the intestine to fight against viral and parasitic infections. Furthermore, intestinal stem cells (ISCs) are also indispensable in intestinal immunity. Only the coordinated combination of the intestinal immune defense system and intestinal tissue renewal can effectively defend against pathogenic microorganisms.
Collapse
|
18
|
Zhou H, Wu S, Liu L, Liu X, Lan S, Jiang J, Yang W, Jin P, Xia X, Ma F. Drosophila Relish-mediated miR-317 expression facilitates immune homeostasis restoration via inhibiting PGRP-LC. Eur J Immunol 2022; 52:1934-1945. [PMID: 36155909 DOI: 10.1002/eji.202250034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 12/13/2022]
Abstract
Innate immunity is the first and essential line for resisting pathogens, and the immune intensity and duration need to be strictly regulated to balance excessive or insufficient immune response. MicroRNAs (miRNAs) are crucial regulators of immune response in Drosophila, yet how immune-related miRNAs are regulated remains poorly understood. Herein, we elucidated that the involvement of miR-317 in NF-κB transcription factor Relish mediated Drosophila Imd pathway in response to Gram-negative (G-) bacteria stimulation. Remarkably, the dynamic expression profiling for immune response indicated that Relish simultaneously enhances the expression of the effector antimicrobial peptide Dpt as well as miR-317 post-infection. Upregulation of miR-317 could further down-regulate the expression of PGRP-LC, thereby forming a feedback in Drosophila Imd pathway to prevent over-activation and restore immune homeostasis. Taken together, our study not only uncovers a novel Relish/miR-317/PGRP-LC regulatory axis to attenuate Drosophila Imd immune response and facilitate immune homeostasis restoration, but also provides vital insights into the complex mechanisms of animal innate immune regulation.
Collapse
Affiliation(s)
- Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China.,Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China
| | - Shanshan Wu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Li Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Xiaoqi Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Siyu Lan
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Jiajun Jiang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Wan Yang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Xinyi Xia
- Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| |
Collapse
|
19
|
Ogienko AA, Omelina ES, Bylino OV, Batin MA, Georgiev PG, Pindyurin AV. Drosophila as a Model Organism to Study Basic Mechanisms of Longevity. Int J Mol Sci 2022; 23:11244. [PMID: 36232546 PMCID: PMC9569508 DOI: 10.3390/ijms231911244] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
The spatio-temporal regulation of gene expression determines the fate and function of various cells and tissues and, as a consequence, the correct development and functioning of complex organisms. Certain mechanisms of gene activity regulation provide adequate cell responses to changes in environmental factors. Aside from gene expression disorders that lead to various pathologies, alterations of expression of particular genes were shown to significantly decrease or increase the lifespan in a wide range of organisms from yeast to human. Drosophila fruit fly is an ideal model system to explore mechanisms of longevity and aging due to low cost, easy handling and maintenance, large number of progeny per adult, short life cycle and lifespan, relatively low number of paralogous genes, high evolutionary conservation of epigenetic mechanisms and signalling pathways, and availability of a wide range of tools to modulate gene expression in vivo. Here, we focus on the organization of the evolutionarily conserved signaling pathways whose components significantly influence the aging process and on the interconnections of these pathways with gene expression regulation.
Collapse
Affiliation(s)
- Anna A. Ogienko
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
| | - Evgeniya S. Omelina
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
- Laboratory of Biotechnology, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia
| | - Oleg V. Bylino
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology RAS, 119334 Moscow, Russia
| | - Mikhail A. Batin
- Open Longevity, 15260 Ventura Blvd., Sherman Oaks, Los Angeles, CA 91403, USA
| | - Pavel G. Georgiev
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology RAS, 119334 Moscow, Russia
| | - Alexey V. Pindyurin
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
| |
Collapse
|
20
|
Wang M, Wang Y, Chang M, Wang X, Shi Z, Raikhel AS, Zou Z. Ecdysone signaling mediates the trade-off between immunity and reproduction via suppression of amyloids in the mosquito Aedes aegypti. PLoS Pathog 2022; 18:e1010837. [PMID: 36137163 PMCID: PMC9531809 DOI: 10.1371/journal.ppat.1010837] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/04/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
The balance between immunity and reproduction is essential for many key physiological functions. We report that to maintain an optimal fertility, 20-hydroxyecdysone (20E) and the ecdysone receptor (EcR) downregulate the immune deficiency (IMD) pathway during the post blood meal phase (PBM) of the Aedes aegypti reproductive cycle. RNA interference-mediated depletion of EcR elicited an increased expression of the IMD pathway components, and these mosquitoes were more resistant to infection by Gram-negative bacteria. Moreover, 20E and EcR recruit Pirk-like, the mosquito ortholog of Drosophila melanogaster Pirk. CRISPR-Cas9 knockout of Pirk-like has shown that it represses the IMD pathway by interfering with IMD-mediated formation of amyloid aggregates. 20E and EcR disruption of the amyloid formation is pivotal for maintaining normal yolk protein production and fertility. Additionally, 20E and its receptor EcR directly induce Pirk-like to interfere with cRHIM-mediated formation of amyloid. Our study highlights the vital role of 20E in governing the trade-off between immunity and reproduction. Pirk-like might be a potential target for new methods to control mosquito reproduction and pathogen transmission.
Collapse
Affiliation(s)
- Mao Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yanhong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Mengmeng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xueli Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zuokun Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Alexander S. Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
- * E-mail: (ASR); (ZZ)
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- * E-mail: (ASR); (ZZ)
| |
Collapse
|
21
|
Cammarata-Mouchtouris A, Acker A, Goto A, Chen D, Matt N, Leclerc V. Dynamic Regulation of NF-κB Response in Innate Immunity: The Case of the IMD Pathway in Drosophila. Biomedicines 2022; 10:2304. [PMID: 36140409 PMCID: PMC9496462 DOI: 10.3390/biomedicines10092304] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Metazoans have developed strategies to protect themselves from pathogenic attack. These preserved mechanisms constitute the immune system, composed of innate and adaptive responses. Among the two kinds, the innate immune system involves the activation of a fast response. NF-κB signaling pathways are activated during infections and lead to the expression of timely-controlled immune response genes. However, activation of NF-κB pathways can be deleterious when uncontrolled. Their regulation is necessary to prevent the development of inflammatory diseases or cancers. The similarity of the NF-κB pathways mediating immune mechanisms in insects and mammals makes Drosophila melanogaster a suitable model for studying the innate immune response and learning general mechanisms that are also relevant for humans. In this review, we summarize what is known about the dynamic regulation of the central NF-κB-pathways and go into detail on the molecular level of the IMD pathway. We report on the role of the nuclear protein Akirin in the regulation of the NF-κB Relish immune response. The use of the Drosophila model allows the understanding of the fine-tuned regulation of this central NF-κB pathway.
Collapse
Affiliation(s)
| | - Adrian Acker
- Institut de Biologie Moléculaire et Cellulaire (IBMC), UPR9022, CNRS, Université de Strasbourg, 67084 Strasbourg, France
| | - Akira Goto
- Institut de Biologie Moléculaire et Cellulaire (IBMC), UPR9022, CNRS, Université de Strasbourg, 67084 Strasbourg, France
| | - Di Chen
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Nicolas Matt
- Institut de Biologie Moléculaire et Cellulaire (IBMC), UPR9022, CNRS, Université de Strasbourg, 67084 Strasbourg, France
| | - Vincent Leclerc
- Institut de Biologie Moléculaire et Cellulaire (IBMC), UPR9022, CNRS, Université de Strasbourg, 67084 Strasbourg, France
| |
Collapse
|
22
|
Dai M, Yang J, Liu X, Gu H, Li F, Li B, Wei J. Parasitism by the Tachinid Parasitoid Exorista japonica Leads to Suppression of Basal Metabolism and Activation of Immune Response in the Host Bombyx mori. INSECTS 2022; 13:insects13090792. [PMID: 36135493 PMCID: PMC9506100 DOI: 10.3390/insects13090792] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 05/26/2023]
Abstract
The dipteran tachinid parasitoids are important biocontrol agents, and they must survive the harsh environment and rely on the resources of the host insect to complete their larval stage. We have previously demonstrated that the parasitism by the tachinid parasitoid Exoristajaponica, a pest of the silkworm, causes pupation defects in Bombyx mori. However, the underlying mechanism is not fully understood. Here, we performed transcriptome analysis of the fat body of B. mori parasitized by E. japonica. We identified 1361 differentially expressed genes, with 394 genes up-regulated and 967 genes down-regulated. The up-regulated genes were mainly associated with immune response, endocrine system and signal transduction, whereas the genes related to basal metabolism, including energy metabolism, transport and catabolism, lipid metabolism, amino acid metabolism and carbohydrate metabolism were down-regulated, indicating that the host appeared to be in poor nutritional status but active in immune response. Moreover, by time-course gene expression analysis we found that genes related to amino acid synthesis, protein degradation and lipid metabolism in B. mori at later parasitization stages were inhibited. Antimicrobial peptides including Cecropin A, Gloverin and Moricin, and an immulectin, CTL11, were induced. These results indicate that the tachinid parasitoid perturbs the basal metabolism and induces the energetically costly immunity of the host, and thus leading to incomplete larval-pupal ecdysis of the host. This study provided insights into how tachinid parasitoids modify host basal metabolism and immune response for the benefit of developing parasitoid larvae.
Collapse
Affiliation(s)
- Minli Dai
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Jin Yang
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Xinyi Liu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Haoyi Gu
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Fanchi Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Sericulture Institute, Soochow University, Suzhou 215123, China
| | - Bing Li
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Sericulture Institute, Soochow University, Suzhou 215123, China
| | - Jing Wei
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
- Sericulture Institute, Soochow University, Suzhou 215123, China
- College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
23
|
Yoo TJ, Sup Shim M, Bang J, Kim JH, Jae Lee B. SPS1 deficiency-triggered PGRP-LC and Toll expression controls innate immunity in Drosophila S2 cells. Biol Open 2022; 11:275744. [PMID: 35723425 PMCID: PMC9364239 DOI: 10.1242/bio.059295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/14/2022] [Indexed: 12/29/2022] Open
Abstract
Selenophosphate synthetase 1 (SPS1) is an essential gene for the cell growth and embryogenesis in Drosophila melanogaster. We have previously reported that SPS1 deficiency stimulates the expression of genes responsible for the innate immune system, including antimicrobial peptides (AMPs), in Drosophila S2 cells. However, the underlying mechanism has not been elucidated. Here, we investigated the immune pathways that control the SPS1-deficiency-induced expression of AMPs in S2 cells. It was found that the activation of AMP expression is regulated by both immune deficiency (IMD) and the Toll pathway. Double knockdown of the upstream genes of each pathway with SPS1 showed that the peptidoglycan recognition protein-LC (PGRP-LC) and Toll genes are targeted by SPS1 for regulating these pathways. We also found that the IMD and Toll pathway regulate AMP expression by cross-talking. The levels of PGRP-LC and Toll mRNAs were upregulated upon Sps1 knockdown (6.4±0.36 and 3.2±0.45-fold, respectively, n=3). Overexpression of each protein also upregulated AMPs. Interestingly, PGRP-LC overexpression upregulated AMP more than Toll overexpression. These data strongly suggest that SPS1 controls the innate immune system of D. melanogaster through regulating PGRP-LC and Toll expression.
Collapse
Affiliation(s)
- Tack-Jin Yoo
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Myoung Sup Shim
- Department of Ophthalmology, Duke Eye Center, Duke Eye Center, Duke University, Durham, NC 27705, USA
| | - Jeyoung Bang
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Jin-Hong Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Byeong Jae Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Korea,Author for correspondence ()
| |
Collapse
|
24
|
Hua Y, Zhu Y, Hu Y, Kong F, Duan R, Zhang C, Zhang C, Zhang S, Jin Y, Ye Y, Cai Q, Ji S. A Feedback Regulatory Loop Involving dTrbd/dTak1 in Controlling IMD Signaling in Drosophila Melanogaster. Front Immunol 2022; 13:932268. [PMID: 35911722 PMCID: PMC9329959 DOI: 10.3389/fimmu.2022.932268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/23/2022] [Indexed: 11/23/2022] Open
Abstract
Negative regulators of the inflammatory responses are essential for the maintenance of immune homeostasis and organismal fitness. In Drosophila, the deubiquitinase (Dub) dTrbd selectively restricts the K63-linked ubiquitination modification of dTak1, a pivotal kinase of the IMD signaling pathway, to regulate the IMD innate immune response. However, which domain and how it functions to enable dTrbd's activity remain unexplored. Here, we provide compelling evidence showing that the NZF domain of dTrbd is essential for its association with dTak1. Meanwhile, the Linker region of dTrbd is involved in modulating its condensation, a functional state representing the Dub enzymatical activity of dTrbd. Of interest, the activated IMD signals following bacterial stimuli enhance the dTrbd/dTak1 interaction, as well as the condensate assembly and Dub enzymatical activity of dTrbd. Collectively, our studies shed light on the dual mechanisms by which the IMD signaling-mediated feedback loop of dTrbd/dTak1 precisely regulates the innate immune response in Drosophila.
Collapse
Affiliation(s)
- Yongzhi Hua
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Yangyang Zhu
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Yixuan Hu
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Fanrui Kong
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Renjie Duan
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, China
| | - Chao Zhang
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Chuchu Zhang
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Shikun Zhang
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Yiheng Jin
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Yizhu Ye
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Qingshuang Cai
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Shanming Ji
- Centre for Developmental Biology, School of Life Sciences, Anhui Agricultural University, Hefei, China
| |
Collapse
|
25
|
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: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune response provides the first line of defense against invading pathogens, and immune disorders cause a variety of diseases. The fruit fly Drosophila melanogaster employs multiple innate immune reactions to resist infection. First, epithelial tissues function as physical barriers to prevent pathogen invasion. In addition, macrophage-like plasmatocytes eliminate intruders through phagocytosis, and lamellocytes encapsulate large particles, such as wasp eggs, that cannot be phagocytosed. Regarding humoral immune responses, the fat body, equivalent to the mammalian liver, secretes antimicrobial peptides into hemolymph, killing bacteria and fungi. Drosophila has been shown to be a powerful in vivo model for studying the mechanism of innate immunity and host-pathogen interactions because Drosophila and higher organisms share conserved signaling pathways and factors. Moreover, the ease with which Drosophila genetic and physiological characteristics can be manipulated prevents interference by adaptive immunity. In this review, we discuss the signaling pathways activated in Drosophila innate immunity, namely, the Toll, Imd, JNK, JAK/STAT pathways, and other factors, as well as relevant regulatory networks. We also review the mechanisms by which different tissues, including hemocytes, the fat body, the lymph gland, muscles, the gut and the brain coordinate innate immune responses. Furthermore, the latest studies in this field are outlined in this review. In summary, understanding the mechanism underlying innate immunity orchestration in Drosophila will help us better study human innate immunity-related diseases.
Collapse
|
26
|
Mutations of γCOP Gene Disturb Drosophila melanogaster Innate Immune Response to Pseudomonas aeruginosa. Int J Mol Sci 2022; 23:ijms23126499. [PMID: 35742941 PMCID: PMC9223523 DOI: 10.3390/ijms23126499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Drosophila melanogaster (the fruit fly) is a valuable experimental platform for modeling host–pathogen interactions. It is also commonly used to define innate immunity pathways and to understand the mechanisms of both host tolerance to commensal microbiota and response to pathogenic agents. Herein, we investigate how the host response to bacterial infection is mirrored in the expression of genes of Imd and Toll pathways when D. melanogaster strains with different γCOP genetic backgrounds are infected with Pseudomonas aeruginosa ATCC 27853. Using microarray technology, we have interrogated the whole-body transcriptome of infected versus uninfected fruit fly males with three specific genotypes, namely wild-type Oregon, γCOPS057302/TM6B and γCOP14a/γCOP14a. While the expression of genes pertaining to Imd and Toll is not significantly modulated by P. aeruginosa infection in Oregon males, many of the components of these cascades are up- or downregulated in both infected and uninfected γCOPS057302/TM6B and γCOP14a/γCOP14a males. Thus, our results suggest that a γCOP genetic background modulates the gene expression profiles of Imd and Toll cascades involved in the innate immune response of D. melanogaster, inducing the occurrence of immunological dysfunctions in γCOP mutants.
Collapse
|
27
|
Zhou H, Wu S, Liu L, Li R, Jin P, Li S. Drosophila Relish Activating lncRNA-CR33942 Transcription Facilitates Antimicrobial Peptide Expression in Imd Innate Immune Response. Front Immunol 2022; 13:905899. [PMID: 35720331 PMCID: PMC9201911 DOI: 10.3389/fimmu.2022.905899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 12/29/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are an emerging class of regulators that play crucial roles in regulating the strength and duration of innate immunity. However, little is known about the regulation of Drosophila innate immunity-related lncRNAs. In this study, we first revealed that overexpression of lncRNA-CR33942 could strengthen the expression of the Imd pathway antimicrobial peptide (AMP) genes Diptericin (Dpt) and Attacin-A (AttA) after infection, and vice versa. Secondly, RNA-seq analysis of lncRNA-CR33942-overexpressing flies post Gram-negative bacteria infection confirmed that lncRNA-CR33942 positively regulated the Drosophila immune deficiency (Imd) pathway. Mechanistically, we found that lncRNA-CR33942 interacts and enhances the binding of NF-κB transcription factor Relish to Dpt and AttA promoters, thereby facilitating Dpt and AttA expression. Relish could also directly promote lncRNA-CR33942 transcription by binding to its promoter. Finally, rescue experiments and dynamic expression profiling post-infection demonstrated the vital role of the Relish/lncRNA-CR33942/AMP regulatory axis in enhancing Imd pathway and maintaining immune homeostasis. Our study elucidates novel mechanistic insights into the role of lncRNA-CR33942 in activating Drosophila Imd pathway and the complex regulatory interaction during the innate immune response of animals.
Collapse
Affiliation(s)
- Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shanshan Wu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Li Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ruimin Li
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shengjie Li
- Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, School of Food Science, Nanjing Xiaozhuang University, Nanjing, China
| |
Collapse
|
28
|
Defining the mechanisms of action and mosquito larva midgut response to a yeast-encapsulated orange oil larvicide. Parasit Vectors 2022; 15:183. [PMID: 35643588 PMCID: PMC9148471 DOI: 10.1186/s13071-022-05307-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Yeast-encapsulated orange oil (YEOO) is a novel, ingestible larvicide that combines the benefits of a low-cost essential oil with yeast, an attractive food source for mosquito larvae. In this work, we investigated the underlying mechanisms of action associated with YEOO ingestion by Aedes aegypti larvae. METHODS Aedes aegypti third-stage larvae (L3) were treated with sublethal or lethal concentrations of YEOO. Genes associated with apoptosis, autophagy and innate immune responses were investigated by RT-qPCR in guts and carcasses dissected from treated and control larvae. Differential expression of cytochrome P450 genes in the CYP6 and CYP9 families were also investigated. Confocal and transmission electron microscopy were used to assess damage caused by YEOO throughout the larval alimentary canal. TUNEL was used to assess apoptosis via DNA fragmentation. RESULTS The apoptosis genes IAP1 and IAP2 in larvae displayed opposing effects following exposure to lethal doses of YEOO, with a 26-fold induction of IAP1 at 8 h post YEOO ingestion. The effector caspase CASPS8 displayed a 6.7-fold induction in the gut and concomitant 70-fold induction in the carcass at 8 h post YEOO ingestion. The midgut epithelia regenerator, Vein, had an 11-fold induction in the gut after 4 h and was repressed 7.6-fold in the carcass at 24 h. Sublethal concentrations (< LC50) led to significant differential expression of CYP6 and CYP9 genes. Midgut epithelial damage was highlighted by the destruction of microvilli, vacuolization of midgut cells and damage to cell junctions and basal lamina as early as 30 min. Larval type 2 peritrophic matrix structural integrity and porosity remain unchanged. CONCLUSION Our results strongly suggest that the robust larvicidal activity of YEOO is due to a generalized broad-acting mechanism combining epithelial damage and apoptosis, with concomitant expression of multiple innate response genes involved in epithelial regeneration and detoxification. YEOO's amenability for use as part of an integrated vector management program makes this novel larvicide a practical approach for mosquito larval control in the future.
Collapse
|
29
|
Arias-Rojas A, Iatsenko I. The Role of Microbiota in Drosophila melanogaster Aging. FRONTIERS IN AGING 2022; 3:909509. [PMID: 35821860 PMCID: PMC9261426 DOI: 10.3389/fragi.2022.909509] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/22/2022] [Indexed: 12/24/2022]
Abstract
Intestinal microbial communities participate in essential aspects of host biology, including nutrient acquisition, development, immunity, and metabolism. During host aging, dramatic shifts occur in the composition, abundance, and function of the gut microbiota. Although such changes in the microbiota are conserved across species, most studies remain descriptive and at most suggest a correlation between age-related pathology and particular microbes. Therefore, the causal role of the microbiota in host aging has remained a challenging question, in part due to the complexity of the mammalian intestinal microbiota, most of which is not cultivable or genetically amenable. Here, we summarize recent studies in the fruit fly Drosophila melanogaster that have substantially progressed our understanding at the mechanistic level of how gut microbes can modulate host aging.
Collapse
Affiliation(s)
| | - Igor Iatsenko
- Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
30
|
Soory A, Ratnaparkhi GS. SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 2022; 18:e1010356. [PMID: 35255103 PMCID: PMC8929699 DOI: 10.1371/journal.ppat.1010356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 03/17/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Post-translational modification by the small ubiquitin-like modifier, SUMO can modulate the activity of its conjugated proteins in a plethora of cellular contexts. The effect of SUMO conjugation of proteins during an immune response is poorly understood in Drosophila. We have previously identified that the transcription factor Jra, the Drosophila Jun ortholog and a member of the AP-1 complex is one such SUMO target. Here, we find that Jra is a regulator of the Pseudomonas entomophila induced gut immune gene regulatory network, modulating the expression of a few thousand genes, as measured by quantitative RNA sequencing. Decrease in Jra in gut enterocytes is protective, suggesting that reduction of Jra signaling favors the host over the pathogen. In Jra, lysines 29 and 190 are SUMO conjugation targets, with the JraK29R+K190R double mutant being SUMO conjugation resistant (SCR). Interestingly, a JraSCR fly line, generated by CRISPR/Cas9 based genome editing, is more sensitive to infection, with adults showing a weakened host response and increased proliferation of Pseudomonas. Transcriptome analysis of the guts of JraSCR and JraWT flies suggests that lack of SUMOylation of Jra significantly changes core elements of the immune gene regulatory network, which include antimicrobial agents, secreted ligands, feedback regulators, and transcription factors. Mechanistically, SUMOylation attenuates Jra activity, with the TFs, forkhead, anterior open, activating transcription factor 3 and the master immune regulator Relish being important transcriptional targets. Our study implicates Jra as a major immune regulator, with dynamic SUMO conjugation/deconjugation of Jra modulating the kinetics of the gut immune response. The intestine has a resident population of commensal microorganisms against which the immune machinery is tuned to show low or no reactivity. In contrast, when pathogenic microorganisms are ingested, the gut responds by activating signaling cascades that lead to the killing and clearance of the pathogen. In this study, we examine the role played by the well-known transcription factor Jun in regulating the immune response in the Drosophila gut. We find that loss of Jun leads to the change in intensity and kinetics of the gut immune transcriptome. The transcriptional profile indicates a stronger response when Jun activity is reduced. Also, animals infected with Pseudomonas entomophila live longer when Jun signaling is reduced. Further, we find that Jun is post-translationally modified on Lys29 and Lys190 by SUMO. To understand the effect of SUMO-conjugation of Jun, we create by state-of-the-art CRISPR/Cas9 genome editing a Drosophila line where Jun is resistant to SUMOylation. This line is more sensitive to infection, with a weaker host-defense response. Our data suggest that Jun Signaling favors the pathogen by dampening the immune response. SUMO conjugation of Jun reverses the dampening and strengthens the immune response in favor of the host. Dynamic SUMOylation of Jun thus fine-tunes the gut immune response to pathogens.
Collapse
Affiliation(s)
- Amarendranath Soory
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| | - Girish S. Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| |
Collapse
|
31
|
McKenna CH, Asgari D, Crippen TL, Zheng L, Sherman RA, Tomberlin JK, Meisel RP, Tarone AM. Gene expression in Lucilia sericata (Diptera: Calliphoridae) larvae exposed to Pseudomonas aeruginosa and Acinetobacter baumannii identifies shared and microbe-specific induction of immune genes. INSECT MOLECULAR BIOLOGY 2022; 31:85-100. [PMID: 34613655 DOI: 10.1111/imb.12740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 06/13/2023]
Abstract
Antibiotic resistance is a continuing challenge in medicine. There are various strategies for expanding antibiotic therapeutic repertoires, including the use of blow flies. Their larvae exhibit strong antibiotic and antibiofilm properties that alter microbiome communities. One species, Lucilia sericata, is used to treat problematic wounds due to its debridement capabilities and its excretions and secretions that kill some pathogenic bacteria. There is much to be learned about how L. sericata interacts with microbiomes at the molecular level. To address this deficiency, gene expression was assessed after feeding exposure (1 h or 4 h) to two clinically problematic pathogens: Pseudomonas aeruginosa and Acinetobacter baumannii. The results identified immunity-related genes that were differentially expressed when exposed to these pathogens, as well as non-immune genes possibly involved in gut responses to bacterial infection. There was a greater response to P. aeruginosa that increased over time, while few genes responded to A. baumannii exposure, and expression was not time-dependent. The response to feeding on pathogens indicates a few common responses and features distinct to each pathogen, which is useful in improving the wound debridement therapy and helps to develop biomimetic alternatives.
Collapse
Affiliation(s)
- C H McKenna
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - D Asgari
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - T L Crippen
- Southern Plains Agricultural Research Center, Agricultural Research Service, US Department of Agriculture, College Station, TX, USA
| | - L Zheng
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - R A Sherman
- BioTherapeutics, Education and Research (BTER) Foundation, Irvine, CA, USA
- Monarch Labs, Irvine, CA, USA
| | - J K Tomberlin
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - R P Meisel
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - A M Tarone
- Department of Entomology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
32
|
Prigot-Maurice C, Beltran-Bech S, Braquart-Varnier C. Why and how do protective symbionts impact immune priming with pathogens in invertebrates? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104245. [PMID: 34453995 DOI: 10.1016/j.dci.2021.104245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/29/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
Growing evidence demonstrates that invertebrates display adaptive-like immune abilities, commonly known as "immune priming". Immune priming is a process by which a host improves its immune defences following an initial pathogenic exposure, leading to better protection after a subsequent infection with the same - or different - pathogens. Nevertheless, beneficial symbionts can enhance similar immune priming processes in hosts, such as when they face repeated infections with pathogens. This "symbiotic immune priming" protects the host against pathogenic viruses, bacteria, fungi, or eukaryotic parasites. In this review, we explore the extent to which protective symbionts interfere and impact immune priming against pathogens from both a mechanical (proximal) and an evolutionary (ultimate) point of view. We highlight that the immune priming of invertebrates is the cornerstone of the tripartite interaction of hosts/symbionts/pathogens. The main shared mechanism of immune priming (induced by symbionts or pathogens) is the sustained immune response at the beginning of host-microbial interactions. However, the evolutionary outcome of immune priming leads to a specific discrimination, which provides enhanced tolerance or resistance depending on the type of microbe. Based on several studies testing immune priming against pathogens in the presence or absence of protective symbionts, we observed that both types of immune priming could overlap and affect each other inside the same hosts. As protective symbionts could be an evolutionary force that influences immune priming, they may help us to better understand the heterogeneity of pathogenic immune priming across invertebrate populations and species.
Collapse
Affiliation(s)
- Cybèle Prigot-Maurice
- Université de Poitiers - UFR Sciences Fondamentales et Appliquées, Laboratoire Écologie et Biologie des Interactions - UMR CNRS 7267, Bâtiment B8-B35, 5 rue Albert Turpin, TSA 51106, F, 86073, POITIERS Cedex 9, France.
| | - Sophie Beltran-Bech
- Université de Poitiers - UFR Sciences Fondamentales et Appliquées, Laboratoire Écologie et Biologie des Interactions - UMR CNRS 7267, Bâtiment B8-B35, 5 rue Albert Turpin, TSA 51106, F, 86073, POITIERS Cedex 9, France
| | - Christine Braquart-Varnier
- Université de Poitiers - UFR Sciences Fondamentales et Appliquées, Laboratoire Écologie et Biologie des Interactions - UMR CNRS 7267, Bâtiment B8-B35, 5 rue Albert Turpin, TSA 51106, F, 86073, POITIERS Cedex 9, France
| |
Collapse
|
33
|
Rodrigues MA, Merckelbach A, Durmaz E, Kerdaffrec E, Flatt T. Transcriptomic evidence for a trade-off between germline proliferation and immunity in Drosophila. Evol Lett 2021; 5:644-656. [PMID: 34917403 PMCID: PMC8645197 DOI: 10.1002/evl3.261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 11/08/2022] Open
Abstract
Life-history theory posits that investment into reproduction might occur at the expense of investment into somatic maintenance, including immune function. If so, reduced or curtailed reproductive effort might be expected to increase immunity. In support of this notion, work in Caenorhabditis elegans has shown that worms lacking a germline exhibit improved immunity, but whether the antagonistic relation between germline proliferation and immunity also holds for other organisms is less well understood. Here, we report that transgenic ablation of germ cells in late development or early adulthood in Drosophila melanogaster causes elevated baseline expression and increased induction of Toll and Imd immune genes upon bacterial infection, as compared to fertile flies with an intact germline. We also identify immune genes whose expression after infection differs between fertile and germline-less flies in a manner that is conditional on their mating status. We conclude that germline activity strongly impedes the expression and inducibility of immune genes and that this physiological trade-off might be evolutionarily conserved.
Collapse
Affiliation(s)
| | | | - Esra Durmaz
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Envel Kerdaffrec
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Thomas Flatt
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| |
Collapse
|
34
|
Salem Wehbe L, Barakat D, Acker A, El Khoury R, Reichhart JM, Matt N, El Chamy L. Protein Phosphatase 4 Negatively Regulates the Immune Deficiency-NF-κB Pathway during the Drosophila Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1616-1626. [PMID: 34452932 PMCID: PMC7616922 DOI: 10.4049/jimmunol.1901497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/07/2021] [Indexed: 12/31/2022]
Abstract
The evolutionarily conserved immune deficiency (IMD) signaling pathway shields Drosophila against bacterial infections. It regulates the expression of antimicrobial peptides encoding genes through the activation of the NF-κB transcription factor Relish. Tight regulation of the signaling cascade ensures a balanced immune response, which is otherwise highly harmful. Several phosphorylation events mediate intracellular progression of the IMD pathway. However, signal termination by dephosphorylation remains largely elusive. Here, we identify the highly conserved protein phosphatase 4 (PP4) complex as a bona fide negative regulator of the IMD pathway. RNA interference-mediated gene silencing of PP4-19c, PP4R2, and Falafel, which encode the catalytic and regulatory subunits of the phosphatase complex, respectively, caused a marked upregulation of bacterial-induced antimicrobial peptide gene expression in both Drosophila melanogaster S2 cells and adult flies. Deregulated IMD signaling is associated with reduced lifespan of PP4-deficient flies in the absence of any infection. In contrast, flies overexpressing this phosphatase are highly sensitive to bacterial infections. Altogether, our results highlight an evolutionarily conserved function of PP4c in the regulation of NF-κB signaling from Drosophila to mammals.
Collapse
Affiliation(s)
- Layale Salem Wehbe
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Dana Barakat
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Adrian Acker
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Rita El Khoury
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | | | - Nicolas Matt
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Laure El Chamy
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| |
Collapse
|
35
|
Disparate regulation of IMD signaling drives sex differences in infection pathology in Drosophila melanogaster. Proc Natl Acad Sci U S A 2021; 118:2026554118. [PMID: 34341118 PMCID: PMC8364183 DOI: 10.1073/pnas.2026554118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sex differences in infection outcome are a widely observed phenomenon. While it is known that biological sex can influence an animal’s response to infection, the mechanisms through which these differences emerge are less clear. Here, we describe a mechanism through which heightened regulation of the IMD signaling pathway by female—but not male—Drosophila melanogaster reduces the cost of immune activity at the expense of resistance to bacterial infection. Through the masculinization of the main organ responsible for antimicrobial peptide activity in the fly (fat body), this work demonstrates that this heightened immune regulation is mediated by sex-determining pathways. Male and female animals exhibit differences in infection outcomes. One possible source of sexually dimorphic immunity is the sex-specific costs of immune activity or pathology, but little is known about the independent effects of immune- versus microbe-induced pathology and whether these may differ for the sexes. Here, by measuring metabolic and physiological outputs in Drosophila melanogaster with wild-type and mutant immune responses, we test whether the sexes are differentially impacted by these various sources of pathology and identify a critical regulator of this difference. We find that the sexes exhibit differential immune activity but similar bacteria-derived metabolic pathology. We show that female-specific immune-inducible expression of PGRP-LB, a negative regulator of the immune deficiency (IMD) pathway, enables females to reduce immune activity in response to reductions in bacterial numbers. In the absence of PGRP-LB, females are more resistant to infection, confirming the functional importance of this regulation and suggesting that female-biased immune restriction comes at a cost.
Collapse
|
36
|
Prakash P, Roychowdhury-Sinha A, Goto A. Verloren negatively regulates the expression of IMD pathway dependent antimicrobial peptides in Drosophila. Sci Rep 2021; 11:15549. [PMID: 34330981 PMCID: PMC8324896 DOI: 10.1038/s41598-021-94973-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/16/2021] [Indexed: 11/08/2022] Open
Abstract
Drosophila immune deficiency (IMD) pathway is similar to the human tumor necrosis factor receptor (TNFR) signaling pathway and is preferentially activated by Gram-negative bacterial infection. Recent studies highlighted the importance of IMD pathway regulation as it is tightly controlled by numbers of negative regulators at multiple levels. Here, we report a new negative regulator of the IMD pathway, Verloren (Velo). Silencing of Velo led to constitutive expression of the IMD pathway dependent antimicrobial peptides (AMPs), and Escherichia coli stimulation further enhanced the AMP expression. Epistatic analysis indicated that Velo knock-down mediated AMP upregulation is dependent on the canonical members of the IMD pathway. The immune fluorescent study using overexpression constructs revealed that Velo resides both in the nucleus and cytoplasm, but the majority (~ 75%) is localized in the nucleus. We also observed from in vivo analysis that Velo knock-down flies exhibit significant upregulation of the AMP expression and reduced bacterial load. Survival experiments showed that Velo knock-down flies have a short lifespan and are susceptible to the infection of pathogenic Gram-negative bacteria, P. aeruginosa. Taken together, these data suggest that Velo is an additional new negative regulator of the IMD pathway, possibly acting in both the nucleus and cytoplasm.
Collapse
Affiliation(s)
- Pragya Prakash
- INSERM, Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084, Strasbourg, France
| | | | - Akira Goto
- INSERM, Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084, Strasbourg, France.
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
37
|
Harrison MC, Niño LMJ, Rodrigues MA, Ryll J, Flatt T, Oettler J, Bornberg-Bauer E. Gene Coexpression Network Reveals Highly Conserved, Well-Regulated Anti-Ageing Mechanisms in Old Ant Queens. Genome Biol Evol 2021; 13:6263858. [PMID: 33944936 PMCID: PMC8214412 DOI: 10.1093/gbe/evab093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Evolutionary theories of ageing predict a reduction in selection efficiency with age, a so-called “selection shadow,” due to extrinsic mortality decreasing effective population size with age. Classic symptoms of ageing include a deterioration in transcriptional regulation and protein homeostasis. Understanding how ant queens defy the trade-off between fecundity and lifespan remains a major challenge for the evolutionary theory of ageing. It has often been discussed that the low extrinsic mortality of ant queens, that are generally well protected within the nest by workers and soldiers, should reduce the selection shadow acting on old queens. We tested this by comparing strength of selection acting on genes upregulated in young and old queens of the ant, Cardiocondyla obscurior. In support of a reduced selection shadow, we find old-biased genes to be under strong purifying selection. We also analyzed a gene coexpression network (GCN) with the aim to detect signs of ageing in the form of deteriorating regulation and proteostasis. We find no evidence for ageing. In fact, we detect higher connectivity in old queens indicating increased transcriptional regulation with age. Within the GCN, we discover five highly correlated modules that are upregulated with age. These old-biased modules regulate several antiageing mechanisms such as maintenance of proteostasis, transcriptional regulation, and stress response. We observe stronger purifying selection on central hub genes of these old-biased modules compared with young-biased modules. These results indicate a lack of transcriptional ageing in old C. obscurior queens, possibly facilitated by strong selection at old age and well-regulated antiageing mechanisms.
Collapse
Affiliation(s)
- Mark C Harrison
- Institute for Evolution and Biodiversity, University of Münster, Germany
| | | | | | - Judith Ryll
- Institute for Evolution and Biodiversity, University of Münster, Germany
| | - Thomas Flatt
- Department of Biology, University of Fribourg, Switzerland
| | - Jan Oettler
- Institut für Zoologie/Evolutionsbiologie, University of Regensburg, Germany
| | - Erich Bornberg-Bauer
- Department of Biology, University of Fribourg, Switzerland.,Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tübingen, Germany
| |
Collapse
|
38
|
Sciambra N, Chtarbanova S. The Impact of Age on Response to Infection in Drosophila. Microorganisms 2021; 9:microorganisms9050958. [PMID: 33946849 PMCID: PMC8145649 DOI: 10.3390/microorganisms9050958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/26/2023] Open
Abstract
This review outlines the known cellular pathways and mechanisms involved in Drosophila age-dependent immunity to pathogenic microorganisms such as bacteria and fungi. We discuss the implication of host signaling pathways such as the Toll, Immune Deficiency (IMD), Janus kinase signal transducer and activator of transcription (JAK/STAT), and Insulin/Insulin Growth Factor/Target of Rapamycin (IIS/TOR) on immune function with aging. Additionally, we review the effects that factors such as sexual dimorphism, environmental stress, and cellular physiology exert on age-dependent immunity in Drosophila. We discuss potential tradeoffs between heightened immune function and longevity in the absence of infection, and we provide detailed tables outlining the various assays and pathogens used in the cited studies, as well as the age, sex, and strains of Drosophila used. We also discuss the overlapping effects these pathways and mechanisms have on one another. We highlight the great utility of Drosophila as a model organism and the importance of a greater focus on age-dependent antiviral immunity for future studies.
Collapse
|
39
|
Bai S, Yao Z, Raza MF, Cai Z, Zhang H. Regulatory mechanisms of microbial homeostasis in insect gut. INSECT SCIENCE 2021; 28:286-301. [PMID: 32888254 DOI: 10.1111/1744-7917.12868] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Insects live in incredibly complex environments. The intestinal epithelium of insects is in constant contact with microorganisms, some of which are beneficial and some harmful to the host. Insect gut health and function are maintained through multidimensional mechanisms that can proficiently remove foreign pathogenic microorganisms while effectively maintaining local symbiotic microbial homeostasis. The basic immune mechanisms of the insect gut, such as the dual oxidase-reactive oxygen species (Duox-ROS) system and the immune deficiency (Imd)-signaling pathway, are involved in the maintenance of microbial homeostasis. This paper reviews the role of physical defenses, the Duox-ROS and Imd signaling pathways, the Janus kinase/signal transducers and activators of transcription signaling pathway, and intestinal symbiotic flora in the homeostatic maintenance of the insect gut microbiome.
Collapse
Affiliation(s)
- Shuai Bai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhichao Yao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Fahim Raza
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhaohui Cai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
40
|
Yi C, Lv X, Chen D, Sun B, Guo L, Wang S, Ru Y, Wang H, Zeng Q. Transcriptome analysis of the Macrobrachium nipponense hepatopancreas provides insights into immunoregulation under Aeromonas veronii infection. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111503. [PMID: 33120268 DOI: 10.1016/j.ecoenv.2020.111503] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 06/11/2023]
Abstract
The oriental river prawn Macrobrachium nipponense is a commercially important freshwater shrimp that is widely farmed in China. Aeromonas veronii is a conditional pathogen of farmed shrimp, which has caused huge economic losses to the industry. Therefore, there is urgency to study the host-pathogen interactions between M. nipponense and A. veronii to screen individuals with antimicrobial resistance. In this study, we examined the hepatopancreas of moribund M. nipponense infected with A. veronii and healthy individuals at both the histopathological and transcriptomic levels. We showed that A. veronii infection resulted in tubular necrosis of the M. nipponense hepatopancreas. Such changes likely affect assimilation, storage, and excretion by the hepatopancreas, which could ultimately affect the survival and growth of infected individuals. Among the 61,345 unigenes obtained through RNA sequencing and de novo transcriptome assembly, 232 were differentially expressed between the two groups. KEGG and GO analyses revealed that these differentially expressed genes were implicated in pathways, including PPAR, PI3K/AKT, and AMPK signaling. The results of this study will contribute to an analysis of the immune response of M. nipponense to A. veronii infection at the transcriptomic level. Furthermore, the RNA-seq data generated here provide an important genomic resource for research on M. nipponense in the absence of a reference genome.
Collapse
Affiliation(s)
- Cao Yi
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Xiaoting Lv
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Duanduan Chen
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Bing Sun
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Leifeng Guo
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Shouquan Wang
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Yuanyuan Ru
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Hui Wang
- Department of Aquaculture Research Lab, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian 271018, Shandong, China.
| | - Qifan Zeng
- Ministry of Education, Key Laboratory of Marine Genetics and Breeding, College of Marine Science, Ocean University of China, Qingdao 266003, Shandong, China.
| |
Collapse
|
41
|
Wang X, Zhong Z, Chen X, Hong Z, Lin W, Mu X, Hu X, Zheng H. High-Fat Diets with Differential Fatty Acids Induce Obesity and Perturb Gut Microbiota in Honey Bee. Int J Mol Sci 2021; 22:ijms22020834. [PMID: 33467664 PMCID: PMC7830725 DOI: 10.3390/ijms22020834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/18/2022] Open
Abstract
HFD (high-fat diet) induces obesity and metabolic disorders, which is associated with the alteration in gut microbiota profiles. However, the underlying molecular mechanisms of the processes are poorly understood. In this study, we used the simple model organism honey bee to explore how different amounts and types of dietary fats affect the host metabolism and the gut microbiota. Excess dietary fat, especially palm oil, elicited higher weight gain, lower survival rates, hyperglycemic, and fat accumulation in honey bees. However, microbiota-free honey bees reared on high-fat diets did not significantly change their phenotypes. Different fatty acid compositions in palm and soybean oil altered the lipid profiles of the honey bee body. Remarkably, dietary fats regulated lipid metabolism and immune-related gene expression at the transcriptional level. Gene set enrichment analysis showed that biological processes, including transcription factors, insulin secretion, and Toll and Imd signaling pathways, were significantly different in the gut of bees on different dietary fats. Moreover, a high-fat diet increased the relative abundance of Gilliamella, while the level of Bartonella was significantly decreased in palm oil groups. This study establishes a novel honey bee model of studying the crosstalk between dietary fat, gut microbiota, and host metabolism.
Collapse
|
42
|
Swanson LC, Trujillo EA, Thiede GH, Katzenberger RJ, Shishkova E, Coon JJ, Ganetzky B, Wassarman DA. Survival Following Traumatic Brain Injury in Drosophila Is Increased by Heterozygosity for a Mutation of the NF-κB Innate Immune Response Transcription Factor Relish. Genetics 2020; 216:1117-1136. [PMID: 33109529 PMCID: PMC7768241 DOI: 10.1534/genetics.120.303776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) pathologies are caused by primary and secondary injuries. Primary injuries result from physical damage to the brain, and secondary injuries arise from cellular responses to primary injuries. A characteristic cellular response is sustained activation of inflammatory pathways commonly mediated by nuclear factor-κB (NF-κB) transcription factors. Using a Drosophila melanogaster TBI model, we previously found that the main proximal transcriptional response to primary injuries is triggered by activation of Toll and Imd innate immune response pathways that engage NF-κB factors Dif and Relish (Rel), respectively. Here, we found by mass spectrometry that Rel protein level increased in fly heads at 4-8 hr after TBI. To investigate the necessity of Rel for secondary injuries, we generated a null allele, Reldel , by CRISPR/Cas9 editing. When heterozygous but not homozygous, the Reldel mutation reduced mortality at 24 hr after TBI and increased the lifespan of injured flies. Additionally, the effect of heterozygosity for Reldel on mortality was modulated by genetic background and diet. To identify genes that facilitate effects of Reldel on TBI outcomes, we compared genome-wide mRNA expression profiles of uninjured and injured +/+, +/Reldel , and Reldel /Reldel flies at 4 hr following TBI. Only a few genes changed expression more than twofold in +/Reldel flies relative to +/+ and Reldel /Reldel flies, and they were not canonical innate immune response genes. Therefore, Rel is necessary for TBI-induced secondary injuries but in complex ways involving Rel gene dose, genetic background, diet, and possibly small changes in expression of innate immune response genes.
Collapse
Affiliation(s)
- Laura C Swanson
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Edna A Trujillo
- Department of Chemistry, College of Letters & Science, University of Wisconsin-Madison, Madison, Wisconsin 53706
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Gene H Thiede
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Rebeccah J Katzenberger
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Joshua J Coon
- Department of Chemistry, College of Letters & Science, University of Wisconsin-Madison, Madison, Wisconsin 53706
- National Center for Quantitative Biology of Complex Systems, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Morgridge Institute for Research, Madison, Wisconsin 53706
| | - Barry Ganetzky
- Department of Genetics, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - David A Wassarman
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
43
|
Cell Communications among Microorganisms, Plants, and Animals: Origin, Evolution, and Interplays. Int J Mol Sci 2020; 21:ijms21218052. [PMID: 33126770 PMCID: PMC7663094 DOI: 10.3390/ijms21218052] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/17/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Cellular communications play pivotal roles in multi-cellular species, but they do so also in uni-cellular species. Moreover, cells communicate with each other not only within the same individual, but also with cells in other individuals belonging to the same or other species. These communications occur between two unicellular species, two multicellular species, or between unicellular and multicellular species. The molecular mechanisms involved exhibit diversity and specificity, but they share common basic features, which allow common pathways of communication between different species, often phylogenetically very distant. These interactions are possible by the high degree of conservation of the basic molecular mechanisms of interaction of many ligand-receptor pairs in evolutionary remote species. These inter-species cellular communications played crucial roles during Evolution and must have been positively selected, particularly when collectively beneficial in hostile environments. It is likely that communications between cells did not arise after their emergence, but were part of the very nature of the first cells. Synchronization of populations of non-living protocells through chemical communications may have been a mandatory step towards their emergence as populations of living cells and explain the large commonality of cell communication mechanisms among microorganisms, plants, and animals.
Collapse
|
44
|
Alameh S, Bartolo G, O’Brien S, Henderson EA, Gonzalez LO, Hartmann S, Klimko CP, Shoe JL, Cote CK, Grill LK, Levitin A, Martchenko Shilman M. Anthrax toxin component, Protective Antigen, protects insects from bacterial infections. PLoS Pathog 2020; 16:e1008836. [PMID: 32866212 PMCID: PMC7458312 DOI: 10.1371/journal.ppat.1008836] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 07/24/2020] [Indexed: 01/23/2023] Open
Abstract
Anthrax is a major zoonotic disease of wildlife, and in places like West Africa, it can be caused by Bacillus anthracis in arid nonsylvatic savannahs, and by B. cereus biovar anthracis (Bcbva) in sylvatic rainforests. Bcbva-caused anthrax has been implicated in as much as 38% of mortality in rainforest ecosystems, where insects can enhance the transmission of anthrax-causing bacteria. While anthrax is well-characterized in mammals, its transmission by insects points to an unidentified anthrax-resistance mechanism in its vectors. In mammals, a secreted anthrax toxin component, 83 kDa Protective Antigen (PA83), binds to cell-surface receptors and is cleaved by furin into an evolutionary-conserved PA20 and a pore-forming PA63 subunits. We show that PA20 increases the resistance of Drosophila flies and Culex mosquitoes to bacterial challenges, without directly affecting the bacterial growth. We further show that the PA83 loop known to be cleaved by furin to release PA20 from PA63 is, in part, responsible for the PA20-mediated protection. We found that PA20 binds directly to the Toll activating peptidoglycan-recognition protein-SA (PGRP-SA) and that the Toll/NF-κB pathway is necessary for the PA20-mediated protection of infected flies. This effect of PA20 on innate immunity may also exist in mammals: we show that PA20 binds to human PGRP-SA ortholog. Moreover, the constitutive activity of Imd/NF-κB pathway in MAPKK Dsor1 mutant flies is sufficient to confer the protection from bacterial infections in a manner that is independent of PA20 treatment. Lastly, Clostridium septicum alpha toxin protects flies from anthrax-causing bacteria, showing that other pathogens may help insects resist anthrax. The mechanism of anthrax resistance in insects has direct implications on insect-mediated anthrax transmission for wildlife management, and with potential for applications, such as reducing the sensitivity of pollinating insects to bacterial pathogens.
Collapse
Affiliation(s)
- Saleem Alameh
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Gloria Bartolo
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Summer O’Brien
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Elizabeth A. Henderson
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Leandra O. Gonzalez
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Stella Hartmann
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Christopher P. Klimko
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Jennifer L. Shoe
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Christopher K. Cote
- Bacteriology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Laurence K. Grill
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
| | - Anastasia Levitin
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
- * E-mail: (AL); (MMS)
| | - Mikhail Martchenko Shilman
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, United States of America
- * E-mail: (AL); (MMS)
| |
Collapse
|
45
|
Abstract
The gut microbiome plays a critical role in the health of many animals. Honeybees are no exception, as they host a core microbiome that affects their nutrition and immune function. However, the relationship between the honeybee immune system and its gut symbionts is poorly understood. Here, we explore how the beneficial symbiont Snodgrassella alvi affects honeybee immune gene expression. We show that both live and heat-killed S. alvi protect honeybees from the opportunistic pathogen Serratia marcescens and lead to the expression of host antimicrobial peptides. Honeybee immune genes respond differently to live S. alvi compared to heat-killed S. alvi, the latter causing a more extensive immune expression response. We show a preference for Toll pathway upregulation over the Imd pathway in the presence of both live and heat-killed S. alvi. Finally, we find that live S. alvi aids in clearance of S. marcescens from the honeybee gut, supporting a potential role for the symbiont in colonization resistance. Our results show that colonization by the beneficial symbiont S. alvi triggers a replicable honeybee immune response. These responses may benefit the host and the symbiont, by helping to regulate gut microbial members and preventing overgrowth or invasion by opportunists.
Collapse
Affiliation(s)
- Richard D Horak
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Sean P Leonard
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Nancy A Moran
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
46
|
Li R, Zhou H, Jia C, Jin P, Ma F. Drosophila Myc restores immune homeostasis of Imd pathway via activating miR-277 to inhibit imd/Tab2. PLoS Genet 2020; 16:e1008989. [PMID: 32810129 PMCID: PMC7455005 DOI: 10.1371/journal.pgen.1008989] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/28/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Drosophila Myc (dMyc), as a broad-spectrum transcription factor, can regulate the expression of a large number of genes to control diverse cellular processes, such as cell cycle progression, cell growth, proliferation and apoptosis. However, it remains largely unknown about whether dMyc can be involved in Drosophila innate immune response. Here, we have identified dMyc to be a negative regulator of Drosophila Imd pathway via the loss- and gain-of-function screening. We demonstrate that dMyc inhibits Drosophila Imd immune response via directly activating miR-277 transcription, which further inhibit the expression of imd and Tab2-Ra/b. Importantly, dMyc can improve the survival of flies upon infection, suggesting inhibiting Drosophila Imd pathway by dMyc is vital to restore immune homeostasis that is essential for survival. Taken together, our study not only reports a new dMyc-miR-277-imd/Tab2 axis involved in the negative regulation of Drosophila Imd pathway, and provides a new insight into the complex regulatory mechanism of Drosophila innate immune homeostasis maintenance.
Collapse
Affiliation(s)
- Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Chaolong Jia
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
- * E-mail: (PJ); (FM)
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
- * E-mail: (PJ); (FM)
| |
Collapse
|
47
|
The Tripartite Interaction of Host Immunity- Bacillus thuringiensis Infection-Gut Microbiota. Toxins (Basel) 2020; 12:toxins12080514. [PMID: 32806491 PMCID: PMC7472377 DOI: 10.3390/toxins12080514] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Bacillus thuringiensis (Bt) is an important cosmopolitan bacterial entomopathogen, which produces various protein toxins that have been expressed in transgenic crops. The evolved molecular interaction between the insect immune system and gut microbiota is changed during the Bt infection process. The host immune response, such as the expression of induced antimicrobial peptides (AMPs), the melanization response, and the production of reactive oxygen species (ROS), varies with different doses of Bt infection. Moreover, B. thuringiensis infection changes the abundance and structural composition of the intestinal bacteria community. The activated immune response, together with dysbiosis of the gut microbiota, also has an important effect on Bt pathogenicity and insect resistance to Bt. In this review, we attempt to clarify this tripartite interaction of host immunity, Bt infection, and gut microbiota, especially the important role of key immune regulators and symbiotic bacteria in the Bt killing activity. Increasing the effectiveness of biocontrol agents by interfering with insect resistance and controlling symbiotic bacteria can be important steps for the successful application of microbial biopesticides.
Collapse
|
48
|
Arora S, Ligoxygakis P. Beyond Host Defense: Deregulation of Drosophila Immunity and Age-Dependent Neurodegeneration. Front Immunol 2020; 11:1574. [PMID: 32774336 PMCID: PMC7387716 DOI: 10.3389/fimmu.2020.01574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Age-dependent neurodegenerative disorders are a set of diseases that affect millions of individuals worldwide. Apart from a small subset that are the result of well-defined inherited autosomal dominant gene mutations (e.g., those encoding the β-amyloid precursor protein and presenilins), our understanding of the genetic network that underscores their pathology, remains scarce. Genome-wide association studies (GWAS) especially in Alzheimer's disease patients and research in Parkinson's disease have implicated inflammation and the innate immune response as risk factors. However, even if GWAS etiology points toward innate immunity, untangling cause, and consequence is a challenging task. Specifically, it is not clear whether predisposition to de-regulated immunity causes an inadequate response to protein aggregation (such as amyloid or α-synuclein) or is the direct cause of this aggregation. Given the evolutionary conservation of the innate immune response in Drosophila and humans, unraveling whether hyperactive immune response in glia have a protective or pathological role in the brain could be a potential strategy in combating age-related neurological diseases.
Collapse
Affiliation(s)
- Srishti Arora
- Laboratory of Cell Biology, Development and Genetics, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Petros Ligoxygakis
- Laboratory of Cell Biology, Development and Genetics, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Huot L, Bigourdan A, Pagès S, Ogier JC, Girard PA, Nègre N, Duvic B. Partner-specific induction of Spodoptera frugiperda immune genes in response to the entomopathogenic nematobacterial complex Steinernema carpocapsae-Xenorhabdus nematophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 108:103676. [PMID: 32184079 DOI: 10.1016/j.dci.2020.103676] [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: 01/23/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 06/10/2023]
Abstract
The Steinernema carpocapsae-Xenorhabdus nematophila association is a nematobacterial complex used in biological control of insect crop pests. The infection success of this dual pathogen strongly depends on its interactions with the host's immune system. Here, we used the lepidopteran pest Spodoptera frugiperda to analyze the respective impact of each partner in the induction of its immune responses. First, we used previously obtained RNAseq data to construct the immunome of S. frugiperda and analyze its induction. We then selected representative genes to study by RT-qPCR their induction kinetics and specificity after independent injections of each partner. We showed that both X. nematophila and S. carpocapsae participate in the induction of stable immune responses to the complex. While X. nematophila mainly induces genes classically involved in antibacterial responses, S. carpocapsae induces lectins and genes involved in melanization and encapsulation. We discuss putative relationships between these differential inductions and the pathogen immunosuppressive strategies.
Collapse
Affiliation(s)
- Louise Huot
- DGIMI, Univ Montpellier, INRAE, Montpellier, France
| | | | - Sylvie Pagès
- DGIMI, Univ Montpellier, INRAE, Montpellier, France
| | | | | | - Nicolas Nègre
- DGIMI, Univ Montpellier, INRAE, Montpellier, France.
| | - Bernard Duvic
- DGIMI, Univ Montpellier, INRAE, Montpellier, France.
| |
Collapse
|
50
|
Valanne S, Järvelä-Stölting M, Harjula SKE, Myllymäki H, Salminen TS, Rämet M. Osa-Containing Brahma Complex Regulates Innate Immunity and the Expression of Metabolic Genes in Drosophila. THE JOURNAL OF IMMUNOLOGY 2020; 204:2143-2155. [PMID: 32198143 DOI: 10.4049/jimmunol.1900571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 02/12/2020] [Indexed: 01/20/2023]
Abstract
Negative regulation of innate immunity is essential to avoid autoinflammation. In Drosophila melanogaster, NF-κB signaling-mediated immune responses are negatively regulated at multiple levels. Using a Drosophila RNA interference in vitro screen, we identified a set of genes inhibiting immune activation. Four of these genes encode members of the chromatin remodeling Osa-containing Brahma (BAP) complex. Silencing additional two genes of the BAP complex was shown to have the same phenotype, confirming its role in immune regulation in vitro. In vivo, the knockdown of osa and brahma was shown to enhance the expression of the Toll pathway-mediated antimicrobial peptides when the flies were challenged with Gram-positive bacteria Micrococcus luteus In this setting, osa knockdown had a particularly strong effect on immune effectors that are predominantly activated by the Imd pathway. Accordingly, Drosophila NF-κB Relish expression was increased by osa silencing. These transcriptional changes were associated with enhanced survival from M. luteus + E. faecalis infection. Besides regulating the expression of immune effector genes, osa RNA interference decreased the expression of a large group of genes involved in metabolism, particularly proteolysis. Of note, the expression of the recently characterized, immune-inducible gene Induced by Infection (IBIN) was diminished in osa knockdown flies. Although IBIN has been shown to modulate metabolism upon infection, the expression of selected Osa-regulated metabolism genes was not rescued by overexpressing IBIN. We conclude that the BAP complex regulates expression of genes involved in metabolism at least partially independent or downstream of IBIN Moreover, Osa affects the NF-κB-mediated immune response by regulating Drosophila NF-κB factor Relish expression.
Collapse
Affiliation(s)
- Susanna Valanne
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland
| | - Mirva Järvelä-Stölting
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland
| | - Sanna-Kaisa E Harjula
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland
| | - Henna Myllymäki
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland
| | - Tiina S Salminen
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland.,Laboratory of Mito-Immuno-Metabolism, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, Faculty of Medicine and Health Technology, 33014 Tampere University, Tampere, Finland; .,PEDEGO Research Unit, Faculty of Medicine, 90014 University of Oulu, Oulu, Finland.,Medical Research Center Oulu, 90014 University of Oulu, Oulu, Finland; and.,Department of Children and Adolescents, Oulu University Hospital, 90014 University of Oulu, Oulu, Finland
| |
Collapse
|