1
|
Ma L, Liu Y, Sun J, Yang X, He Y, Zhang T, Zhao J, Lu Z, Yan X, Qie X. The synthesis of nitric oxide regulated by JNK pathway in the pea aphid to defend against bacterial infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104315. [PMID: 40274239 DOI: 10.1016/j.ibmb.2025.104315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Compared to other insects, the pea aphid Acyrthosiphon pisum exhibits limited immune responses, particularly due to the absence of many immune genes, including those encoding antimicrobial peptides and key components of the IMD pathway. Prior studies proved that the conserved signaling, Jun N-terminal kinase (JNK) pathway, plays a critical role in the immune system of the pea aphid, and nitric oxide synthase (NOS) is required for the pea aphid's defense against infections. Herein, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway directly regulates the expression of NOS and that the JNK pathway-NOS-NO signal axis is efficient in defending against bacterial infections. The Toll pathway is instrumental for combating bacterial infections, and NO can activate the Toll pathway. The Toll pathway induced by NO regulates the expressions of ROS metabolism, lysosome, and phagocytosis-related genes. NO was identified as a crucial signaling molecule that facilitates communication between the JNK and Toll pathways.
Collapse
Affiliation(s)
- Li Ma
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yaya Liu
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jing Sun
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Xiaorong Yang
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yingying He
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Tingting Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Jingyu Zhao
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xizhong Yan
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Xingtao Qie
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| |
Collapse
|
2
|
Diedrick WA, Kanga LHB, Mallinger R, Pescador M, Elsharkawy I, Zhang Y. Molecular Assessment of Genes Linked to Honeybee Health Fed with Different Diets in Nuclear Colonies. INSECTS 2025; 16:374. [PMID: 40332858 DOI: 10.3390/insects16040374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 05/08/2025]
Abstract
Honeybees are of economic importance not only for honey production, but also for crop pollination, which amounts to USD 20 billion per year in the United States. However, the number of honeybee colonies has declined more than 40% during the last few decades. Although this decline is attributed to a combination of factors (parasites, diseases, pesticides, and nutrition), unlike other factors, the effect of nutrition on honeybee health is not well documented. In this study, we assessed the differential expression of seven genes linked to honeybee health under three different diets. These included immune function genes [Cactus, immune deficiency (IMD), Spaetzle)], genes involved in nutrition, cellular defense, longevity, and behavior (Vitellogenin, Malvolio), a gene involved in energy metabolism (Maltase), and a gene associated with locomotory behavior (Single-minded). The diets included (a) commercial pollen patties and sugar syrup, (b) monofloral (anise hyssop), and (c) polyfloral (marigold, anise hyssop, sweet alyssum, and basil). Over the 2.7-month experimental periods, adult bees in controls fed pollen patties and sugar syrup showed upregulated Cactus (involved in Toll pathway) and IMD (signaling pathway controls antibacterial defense) expression, while their counterparts fed monofloral and polyfloral diets downregulated the expression of these genes. Unlike Cactus and IMD, the gene expression profile of Spaetzle (involved in Toll pathway) did not differ across treatments during the experimental period except that it was significantly downregulated on day 63 and day 84 in bees fed polyfloral diets. The Vitellogenin gene indicated that monofloral and polyfloral diets significantly upregulated this gene and enhanced lifespan, foraging behavior, and immunity in adult bees fed with monofloral diets. The expression of Malvolio (involved in sucrose responsiveness and foraging behavior) was upregulated when food reserves (pollen and nectar) were limited in adult bees fed polyfloral diets. Adult bees fed with monofloral diets significantly upregulated the expression of Maltase (involved in energy metabolisms) compared to their counterparts in control diets to the end of the experimental period. Single-Minded Homolog 2 (involved in locomotory behavior) was also upregulated in adult bees fed pollen patties and sugar syrup compared to their counterparts fed monofloral and polyfloral diets. Thus, the food source significantly affected honeybee health and triggered an up- and downregulation of these genes, which correlated with the health and activities of the honeybee colonies. Overall, we found that the companion crops (monofloral and polyfloral) provided higher nutritional benefits to enhance honeybee health than the pollen patty and sugar syrup used currently by beekeepers. Furthermore, while it has been reported that bees require pollen from diverse sources to maintain a healthy physiology and hive, our data on nuclear colonies indicated that a single-species diet (such as anise hyssop) is nutritionally adequate and better or comparable to polyfloral diets. To the best of our knowledge, this is the first report indicating better nutritional benefits from monofloral diets (anise hyssop) over polyfloral diets for honeybee colonies (nucs) in semi-large-scale experimental runs. Thus, we recommend that the landscape of any apiary include highly nutritious food sources, such as anise hyssop, throughout the season to enhance honeybee health.
Collapse
Affiliation(s)
- Worrel A Diedrick
- Entomology Department, College of Agriculture and Food Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Lambert H B Kanga
- Entomology Department, College of Agriculture and Food Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Rachel Mallinger
- Department of Entomology and Nematology, University of Florida, Gainesville, FL 32608, USA
| | - Manuel Pescador
- Entomology Department, College of Agriculture and Food Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Islam Elsharkawy
- Center for Viticulture and Small Fruits Research, Tallahassee, FL 32307, USA
| | - Yanping Zhang
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
3
|
Brutscher F, Germani F, Hausmann G, Jutz L, Basler K. Activation of the Drosophila innate immune system accelerates growth in cooperation with oncogenic Ras. PLoS Biol 2025; 23:e3003068. [PMID: 40294154 PMCID: PMC12036928 DOI: 10.1371/journal.pbio.3003068] [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: 04/23/2024] [Accepted: 02/13/2025] [Indexed: 04/30/2025] Open
Abstract
Innate immunity in Drosophila acts as an organismal surveillance system for external stimuli or cellular fitness and triggers context-specific responses to fight infections and maintain tissue homeostasis. However, uncontrolled activation of innate immune pathways can be detrimental. In mammals, innate immune signaling is often overactivated in malignant cells and contributes to tumor progression. Drosophila tumor models have been instrumental in the discovery of interactions between pathways that promote tumorigenesis, but little is known about whether and how the Toll innate immune pathway interacts with oncogenes. Here we use a Drosophila epithelial in vivo model to investigate the interplay between Toll signaling and oncogenic Ras. In the absence of oncogenic Ras (RasV12), Toll signaling suppresses differentiation and induces apoptosis. In contrast, in the context of RasV12, cells are protected from cell death and Dorsal promotes cell survival and proliferation to drive hyperplasia. Taken together, we show that the tissue-protective functions of innate immune activity can be hijacked by pre-malignant cells to induce tumorous overgrowth.
Collapse
Affiliation(s)
- Fabienne Brutscher
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Federico Germani
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - George Hausmann
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Lena Jutz
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Cacace A, De Leva G, Di Lelio I, Becchimanzi A. Immune-Related Genes in the Honey Bee Mite Varroa destructor ( Acarina, Parasitidae). INSECTS 2025; 16:356. [PMID: 40332846 DOI: 10.3390/insects16040356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025]
Abstract
Despite its ecological and economic importance, many aspects of Varroa destructor's biology remain poorly understood, particularly its defense mechanisms against pathogens. The limited knowledge of Varroa's immunity has hindered the development of RNA interference (RNAi)-based strategies targeting immune-related genes. In this study, we investigated the immune gene repertoire of V. destructor by querying its NCBI nr protein database and comparing it to model species of ticks (Ixodes scapularis) and mites (Galendromus occidentalis and Tetranychus urticae). Transcription of candidate immune genes was confirmed by analyzing a de novo assembled transcriptome of V. destructor. Our findings reveal that V. destructor shares key immunological traits with ticks, including lysozymes, chitinases, and thioester-containing proteins (TEPs), but also shares the absence of transmembrane peptidoglycan recognition proteins (PGRPs), Gram-negative binding proteins, and several lectin families involved in pathogen recognition. Additionally, Varroa mites, like ticks, lack homologs of crucial immune signaling components, such as the unpaired ligand (JAK/STAT), Eiger (JNK), and multiple elements of the IMD pathway. They also do not encode canonical antimicrobial peptides (AMPs) like defensins but possess putative homologs of ctenidins, AMPs previously identified in spiders and ticks, which may be adopted as a novel genetic readout for immune response in mites. Our findings lay the groundwork for future functional studies on mite immunity and open new avenues for RNAi-based biocontrol strategies targeting immune pathways to enhance Varroa management.
Collapse
Affiliation(s)
- Alfonso Cacace
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Giovanna De Leva
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Ilaria Di Lelio
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| | - Andrea Becchimanzi
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
5
|
Wang HY, Chen YY, Liu CJ, Huang SW, Ho ST. Evaluating the Potential Immunostimulatory Effects of Cryptomeria japonica Leaf Essential Oil on Honey Bees (Apis mellifera). ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70040. [PMID: 39966591 DOI: 10.1002/arch.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/20/2025]
Abstract
This study investigated the effects of Cryptomeria japonica leaf essential oil (CjLEO) on honey bee health, focusing on both toxicity and gene expression modulation. Initial toxicity assessments revealed that high concentrations of CjLEO (75 and 100 ppm) were lethal to honey bees, resulting in complete mortality within a short period. Conversely, a lower concentration of 10 ppm exhibited no significant toxic effects, prompting further investigation into its sublethal impacts. Transcriptome analysis via next-generation sequencing demonstrated that CjLEO at 10 ppm induced significant changes in honey bee gene expression compared to the control group. Principal component analysis (PCA) and differential gene expression (DEG) analysis identified more than 9,000 genes, with notable upregulation of immune-related genes, including hymenoptaecin, abaecin, and apidaecin1. Gene ontology (GO) enrichment analysis indicated that these differentially expressed genes were primarily associated with immune responses, such as defense and innate immune pathways. The chemical composition of CjLEO, characterized by GC-MS, identified 16 compounds, with major components including α-pinene, elemol, α-eudesmol, and kaur-16-ene. These compounds are known for their antimicrobial properties, which likely contribute to the observed immunomodulatory effects. CjLEO at a concentration of 10 ppm enhances honey bee immunity without exhibiting significant toxicity, positioning it a promising candidate for improving honey bee resilience against pathogens. Future research should investigate the mechanisms of immune activation and optimize application methods for practical beekeeping, aiming to improve colony health while reducing dependence on synthetic chemicals.
Collapse
Affiliation(s)
- Hao-Yung Wang
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan
| | - Ying-Yu Chen
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan
| | - Chin-Jung Liu
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan
| | - Shih-Wei Huang
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan
| | - Shang-Tse Ho
- Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan
| |
Collapse
|
6
|
Weber K, Karnik D, Brown LD. Transcriptional induction of the IMD signaling pathway and associated antibacterial activity in the digestive tract of cat fleas (Ctenocephalides felis). Parasit Vectors 2024; 17:546. [PMID: 39736773 DOI: 10.1186/s13071-024-06613-x] [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: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Fleas are insect vectors that transmit several Gram-negative bacterial pathogens acquired by ingesting infected vertebrate blood. To combat foodborne illness, insect midgut epithelial cells are armed with efficient microbial recognition and control systems, such as the immune deficiency (IMD) pathway that regulates the expression of antimicrobial peptides (AMPs). However, despite their medical and veterinary importance, relatively little is known about the IMD signaling pathway and production of AMPs in the digestive tract of cat fleas (Ctenocephalides felis). METHODS In the present study, we measured the expression of target genes comprising the IMD pathway, as well as corresponding AMP transcripts, in the digestive tract of C. felis following exposure to three different species of bacteria: Gram-negative Bartonella henselae (a flea-borne pathogen), Gram-negative Serratia marcescens (a model laboratory species), and Gram-positive Micrococcus luteus (a model laboratory species). Additionally, we examined the antibacterial activity of proteins isolated from the flea digestive tract in vitro following bacterial challenge and at different days post adult emergence to determine if feeding-induced antibacterial activity varies with age. RESULTS In our analysis of C. felis, we observed an increase in the expression of representative IMD pathway genes and associated AMP transcripts, indicating the activation of the IMD pathway. Furthermore, our results revealed that different bacterial species elicit distinct transcriptional profiles of IMD pathway genes, suggesting a species-specific response to bacterial invasion. We found that the gut of C. felis produces antibacterial molecules as a localized defense mechanism. Additionally, we observed that proteins with antimicrobial properties are synthesized as part of local defense mechanisms in the gut, with differential patterns of antibacterial activity related to infection status and age. CONCLUSIONS Our findings provide essential insights into the potential mechanisms by which cat fleas regulate immune responses in their digestive tract against different bacterial species.
Collapse
Affiliation(s)
- Katie Weber
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Dhruva Karnik
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Lisa D Brown
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA.
| |
Collapse
|
7
|
Kwon SY, Chan K, Stofanko M, Chan KH, Badenhorst P. Abrupt-mediated control of ninjurins regulates Drosophila sessile haemocyte compartments. Development 2024; 151:dev202977. [PMID: 39545919 DOI: 10.1242/dev.202977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Macrophage-like cells called haemocytes are key effectors of Drosophila cellular innate immune function. Larval haemocytes exist either in circulation or localize to segmentally repeated sessile haemocyte compartments (SHCs). While numerous functions have been proposed for SHCs, the mechanisms directing haemocytes to them are unclear. Here, we have exploited the developmentally regulated dispersal of SHCs that occurs at pupariation to identify the Abrupt (Ab) transcription factor (TF) and ninjurin cell-adhesion molecules as regulators of haemocyte recruitment to SHCs. We show that larval haemocytes express ninjurins, which are required for targeting haemocytes to SHCs. However, at pupariation, ecdysteroid signalling stimulates Ab expression, which collaborates with TFs, including Blimp-1 and Hr3, to repress ninjurins and disperse haemocytes. We observe that experimental manipulations that antagonize ninjurin function in larval haemocytes cause premature SHC dispersal, while stabilization of ninjurins in haemocytes blocks developmentally regulated SHC remodelling and increases sensitivity to immune challenges. Cumulatively, our data indicate that control of ninjurin activity provides a common target through which diverse developmental, environmental and immune stimuli can be integrated to control haemocyte dispersal and immune function.
Collapse
Affiliation(s)
- So Yeon Kwon
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Kimberly Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Martin Stofanko
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Ka Hei Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Paul Badenhorst
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
8
|
Ramesh P, Tiwari SK, Kaizer M, Jangra D, Ghosh K, Mandal S, Mandal L. The NF-κB Factor Relish maintains blood progenitor homeostasis in the developing Drosophila lymph gland. PLoS Genet 2024; 20:e1011403. [PMID: 39250509 PMCID: PMC11424005 DOI: 10.1371/journal.pgen.1011403] [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: 02/06/2024] [Revised: 09/25/2024] [Accepted: 08/24/2024] [Indexed: 09/11/2024] Open
Abstract
Post-larval hematopoiesis in Drosophila largely depends upon the stockpile of progenitors present in the blood-forming organ/lymph gland of the larvae. During larval stages, the lymph gland progenitors gradually accumulate reactive oxygen species (ROS), which is essential to prime them for differentiation. Studies have shown that ROS triggers the activation of JNK (c-Jun Kinase), which upregulates fatty acid oxidation (FAO) to facilitate progenitor differentiation. Intriguingly, despite having ROS, the entire progenitor pool does not differentiate simultaneously in the late larval stages. Using expression analyses, genetic manipulation and pharmacological approaches, we found that the Drosophila NF-κB transcription factor Relish (Rel) shields the progenitor pool from the metabolic pathway that inducts them into the differentiation program by curtailing the activation of JNK. Although ROS serves as the metabolic signal for progenitor differentiation, the input from ROS is monitored by the developmental signal TAK1, which is regulated by Relish. This developmental circuit ensures that the stockpile of ROS-primed progenitors is not exhausted entirely. Our study sheds light on how, during development, integrating NF-κB-like factors with metabolic pathways seem crucial to regulating cell fate transition during development.
Collapse
Affiliation(s)
- Parvathy Ramesh
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Satish Kumar Tiwari
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Md Kaizer
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Deepak Jangra
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Kaustuv Ghosh
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Sudip Mandal
- Molecular Cell and Developmental Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| | - Lolitika Mandal
- Developmental Genetics Laboratory, Indian Institute of Science Education and Research Mohali (IISER Mohali), Punjab, INDIA
| |
Collapse
|
9
|
Huang DY, Qin JS, Dong RK, Liu SN, Chen N, Yuan DW, Li S, Wang Z, Xia X. Ben-JNK signaling is required for host mortality during Periplaneta fuliginosa densovirus infection. PEST MANAGEMENT SCIENCE 2024; 80:4495-4504. [PMID: 38676657 DOI: 10.1002/ps.8154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Cockroaches are widely acknowledged as significant vectors of pathogenic microorganisms. The Periplaneta fuliginosa densovirus (PfDNV) infects the smoky-brown cockroach P. fuliginosa and causes host mortality, which identifies the PfDNV as a species-specific and environmentally friendly biopesticide. However, although the biochemical characterization of PfDNV has been extensively studied, the immune response against PfDNV remains largely unclear. RESULTS Here, we investigated the replication of PfDNV and its associated pathological phenotype in the foregut and hindgut. Consequently, we dissected and performed transcriptome sequencing on the foregut, midgut, and hindgut separately. We revealed the up-regulation of immune response signaling pathway c-Jun N-terminal kinase (JNK) and apoptosis in response to viral infection. Furthermore, knockdown of the JNK upstream gene Ben resulted in a decrease in virus titer and delayed host mortality. CONCLUSION Taken together, our findings provide evidence that the Ben-JNK signaling plays a crucial role in PfDNV infection, leading to excessive apoptosis in intestinal tissues and ultimately resulting in the death of the host. Our results indicated that the host response to PfDNV fosters viral infection, thereby increasing host lethality. This underscores the potential of PfDNV as a viable, environmentally friendly biopesticide. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Dan-Yan Huang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jia-Si Qin
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Ren-Ke Dong
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Su-Ning Liu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Nan Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Dong-Wei Yuan
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou, China
| | - Zhaowei Wang
- State Key Laboratory of Biocontrol, School of Ecology, Sun Yat-sen University, Shenzhen, China
| | - Xiaoling Xia
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
10
|
Brantley SE, Stouthamer CM, Kr P, Fischer ML, Hill J, Schlenke TA, Mortimer NT. Host JAK-STAT activity is a target of parasitoid wasp virulence strategies. PLoS Pathog 2024; 20:e1012349. [PMID: 38950076 PMCID: PMC11244843 DOI: 10.1371/journal.ppat.1012349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/12/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Innate immune responses that allow hosts to survive infection depend on the action of multiple conserved signaling pathways. Pathogens and parasites in turn have evolved virulence factors to target these immune signaling pathways in an attempt to overcome host immunity. Consequently, the interactions between host immune molecules and pathogen virulence factors play an important role in determining the outcome of an infection. The immune responses of Drosophila melanogaster provide a valuable model to understand immune signaling and host-pathogen interactions. Flies are commonly infected by parasitoid wasps and mount a coordinated cellular immune response following infection. This response is characterized by the production of specialized blood cells called lamellocytes that form a tight capsule around wasp eggs in the host hemocoel. The conserved JAK-STAT signaling pathway has been implicated in lamellocyte proliferation and is required for successful encapsulation of wasp eggs. Here we show that activity of Stat92E, the D. melanogaster STAT ortholog, is induced in immune tissues following parasitoid infection. Virulent wasp species are able to suppress Stat92E activity during infection, suggesting they target JAK-STAT pathway activation as a virulence strategy. Furthermore, two wasp species (Leptopilina guineaensis and Ganaspis xanthopoda) suppress phenotypes associated with a gain-of-function mutation in hopscotch, the D. melanogaster JAK ortholog, indicating that they inhibit the activity of the core signaling components of the JAK-STAT pathway. Our data suggest that parasitoid wasp virulence factors block JAK-STAT signaling to overcome fly immune defenses.
Collapse
Affiliation(s)
- Susanna E Brantley
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Corinne M Stouthamer
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Pooja Kr
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Mary L Fischer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Joshua Hill
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
| | - Todd A Schlenke
- Department of Entomology, University of Arizona, Tucson, Arizona, United States of America
| | - Nathan T Mortimer
- School of Biological Sciences, Illinois State University, Normal, Illinois, United States of America
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
11
|
Das BK, Panda SP, Pradhan SP, Raut SS, Kumari M, Meena DK. Molecular insights into STAT1a protein in rohu ( Labeo rohita): unveiling expression profiles, SRC homology domain recognition, and protein-protein interactions triggered by poly I: C. Front Immunol 2024; 15:1398955. [PMID: 38994355 PMCID: PMC11237311 DOI: 10.3389/fimmu.2024.1398955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/09/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction STAT1a is an essential signal transduction protein involved in the interferon pathway, playing a vital role in IFN-alpha/beta and gamma signaling. Limited information is available about the STAT protein in fish, particularly in Indian major carps (IMC). This study aimed to identify and characterize the STAT1a protein in Labeo rohita (LrSTAT1a). Methods The full-length CDS of LrSTAT1a transcript was identified and sequenced. Phylogenetic analyses were performed based on the nucleotide sequences. The in-vivo immune stimulant poly I: C was used to treat various tissues, and the expression of LrSTAT1a was determined using quantitative real-time polymerase chain reaction (qRT-PCR). A 3D model of the STAT1a protein was generated using close structure homologs available in the database and checked using molecular dynamics (MD) simulations. Results The full-length CDS of Labeo rohita STAT1a (LrSTAT1a) transcript consisted of 3238 bp that encoded a polypeptide of 721 amino acids sequence was identified. Phylogenetic analyses were performed based on the nucleotide sequences. Based on our findings, other vertebrates share a high degree of conservation with STAT1a. Additionally, we report that the in vivo immune stimulant poly I: C treatment of various tissues resulted in the expression of LrSTAT1a as determined by quantitative real-time polymerase chain reaction (qRT-PCR). In the current investigation, treatment with poly I: C dramatically increased the expression of LrSTAT1a in nearly every organ and tissue, with the brain, muscle, kidney, and intestine showing the highest levels of expression compared to the control. We made a 3D model of the STAT1a protein by using close structure homologs that were already available in the database. The model was then checked using molecular dynamics (MD) simulations. Consistent with previous research, the MD study highlighted the significance of the STAT1a protein, which is responsible for Src homology 2 (SH2) recognition. An important H-bonding that successfully retains SH2 inside the STAT1a binding cavity was determined to be formed by the conserved residues SER107, GLN530, SER583, LYS584, MET103, and ALA106. Discussion This study provides molecular insights into the STAT1a protein in Rohu (Labeo rohita) and highlights the potential role of STAT1a in the innate immune response in fish. The high degree of conservation of STAT1a among other vertebrates suggests its crucial role in the immune response. The in-vivo immune stimulation results indicate that STAT1a is involved in the immune response in various tissues, with the brain, muscle, kidney, and intestine being the most responsive. The 3D model and MD study provide further evidence of the significance of STAT1a in the immune response, specifically in SH2 recognition. Further research is necessary to understand the specific mechanisms involved in the IFN pathway and the role of STAT1a in the immune response of IMC.
Collapse
Affiliation(s)
- Basanta Kumar Das
- Aquatic Environmental Biotechnology (AEB) Division, Indian Council of Agricultural Research (ICAR) - Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Soumya Prasad Panda
- Aquatic Environmental Biotechnology (AEB) Division, Indian Council of Agricultural Research (ICAR) - Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Smruti Priyambada Pradhan
- Aquatic Environmental Biotechnology (AEB) Division, Indian Council of Agricultural Research (ICAR) - Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Subhashree Subhasmita Raut
- Aquatic Environmental Biotechnology (AEB) Division, Indian Council of Agricultural Research (ICAR) - Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Mala Kumari
- Riverine & Estuaries Fisheries Division, Indian Council of Agricultural Research (ICAR) -Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| | - Dharmendra Kumar Meena
- Open Water Aquaculture Production and Management (OWAPM) Division, Indian Council of Agricultural Research (ICAR)-Central Inland Fisheries Research Institute, Kolkata, West Bengal, India
| |
Collapse
|
12
|
Meraj S, Dhari AS, Mohr E, Lowenberger C, Gries G. A novel prolixicin identified in common bed bugs with activity against both bacteria and parasites. Sci Rep 2024; 14:13818. [PMID: 38879638 PMCID: PMC11180110 DOI: 10.1038/s41598-024-64691-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/12/2024] [Indexed: 06/19/2024] Open
Abstract
The hematophagous common bed bug, Cimex lectularius, is not known to transmit human pathogens outside laboratory settings, having evolved various immune defense mechanisms including the expression of antimicrobial peptides (AMPs). We unveil three novel prolixicin AMPs in bed bugs, exhibiting strong homology to the prolixicin of kissing bugs, Rhodnius prolixus, and to diptericin/attacin AMPs. We demonstrate for the first time sex-specific and immune mode-specific upregulation of these prolixicins in immune organs, the midgut and rest of body, following injection and ingestion of Gr+ (Bacillus subtilis) and Gr- (Escherichia coli) bacteria. Synthetic CL-prolixicin2 significantly inhibited growth of E. coli strains and killed or impeded Trypanosoma cruzi, the Chagas disease agent. Our findings suggest that prolixicins are regulated by both IMD and Toll immune pathways, supporting cross-talk and blurred functional differentiation between major immune pathways. The efficacy of CL-prolixicin2 against T. cruzi underscores the potential of AMPs in Chagas disease management.
Collapse
Affiliation(s)
- Sanam Meraj
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A1S6, Canada.
| | - Arshvir Singh Dhari
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A1S6, Canada
| | - Emerson Mohr
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A1S6, Canada
| | - Carl Lowenberger
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A1S6, Canada
| | - Gerhard Gries
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A1S6, Canada
| |
Collapse
|
13
|
Becchimanzi A, Nicoletti R, Di Lelio I, Russo E. Immune Gene Repertoire of Soft Scale Insects (Hemiptera: Coccidae). Int J Mol Sci 2024; 25:4922. [PMID: 38732132 PMCID: PMC11084805 DOI: 10.3390/ijms25094922] [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: 03/30/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Insects possess an effective immune system, which has been extensively characterized in several model species, revealing a plethora of conserved genes involved in recognition, signaling, and responses to pathogens and parasites. However, some taxonomic groups, characterized by peculiar trophic niches, such as plant-sap feeders, which are often important pests of crops and forestry ecosystems, have been largely overlooked regarding their immune gene repertoire. Here we annotated the immune genes of soft scale insects (Hemiptera: Coccidae) for which omics data are publicly available. By using immune genes of aphids and Drosophila to query the genome of Ericerus pela, as well as the transcriptomes of Ceroplastes cirripediformis and Coccus sp., we highlight the lack of peptidoglycan recognition proteins, galectins, thaumatins, and antimicrobial peptides in Coccidae. This work contributes to expanding our knowledge about the evolutionary trajectories of immune genes and offers a list of promising candidates for developing new control strategies based on the suppression of pests' immunity through RNAi technologies.
Collapse
Affiliation(s)
- Andrea Becchimanzi
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.B.); (I.D.L.); (E.R.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| | - Rosario Nicoletti
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.B.); (I.D.L.); (E.R.)
- Research Centre for Olive, Fruit and Citrus Crops, Council for Agricultural Research and Economics, 81100 Caserta, Italy
| | - Ilaria Di Lelio
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.B.); (I.D.L.); (E.R.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| | - Elia Russo
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.B.); (I.D.L.); (E.R.)
| |
Collapse
|
14
|
Meraj S, Salcedo-Porras N, Lowenberger C, Gries G. Activation of immune pathways in common bed bugs, Cimex lectularius, in response to bacterial immune challenges - a transcriptomics analysis. Front Immunol 2024; 15:1384193. [PMID: 38694504 PMCID: PMC11061471 DOI: 10.3389/fimmu.2024.1384193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
The common bed bug, Cimex lectularius, is an urban pest of global health significance, severely affecting the physical and mental health of humans. In contrast to most other blood-feeding arthropods, bed bugs are not major vectors of pathogens, but the underlying mechanisms for this phenomenon are largely unexplored. Here, we present the first transcriptomics study of bed bugs in response to immune challenges. To study transcriptional variations in bed bugs following ingestion of bacteria, we extracted and processed mRNA from body tissues of adult male bed bugs after ingestion of sterile blood or blood containing the Gram-positive (Gr+) bacterium Bacillus subtilis or the Gram-negative (Gr-) bacterium Escherichia coli. We analyzed mRNA from the bed bugs' midgut (the primary tissue involved in blood ingestion) and from the rest of their bodies (RoB; body minus head and midgut tissues). We show that the midgut exhibits a stronger immune response to ingestion of bacteria than the RoB, as indicated by the expression of genes encoding antimicrobial peptides (AMPs). Both the Toll and Imd signaling pathways, associated with immune responses, were highly activated by the ingestion of bacteria. Bacterial infection in bed bugs further provides evidence for metabolic reconfiguration and resource allocation in the bed bugs' midgut and RoB to promote production of AMPs. Our data suggest that infection with particular pathogens in bed bugs may be associated with altered metabolic pathways within the midgut and RoB that favors immune responses. We further show that multiple established cellular immune responses are preserved and are activated by the presence of specific pathogens. Our study provides a greater understanding of nuances in the immune responses of bed bugs towards pathogens that ultimately might contribute to novel bed bug control tactics.
Collapse
Affiliation(s)
- Sanam Meraj
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | | | | | | |
Collapse
|
15
|
Ma M, Luo J, Li C, Eleftherianos I, Zhang W, Xu L. A life-and-death struggle: interaction of insects with entomopathogenic fungi across various infection stages. Front Immunol 2024; 14:1329843. [PMID: 38259477 PMCID: PMC10800808 DOI: 10.3389/fimmu.2023.1329843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Insects constitute approximately 75% of the world's recognized fauna, with the majority of species considered as pests. Entomopathogenic fungi (EPF) are parasitic microorganisms capable of efficiently infecting insects, rendering them potent biopesticides. In response to infections, insects have evolved diverse defense mechanisms, prompting EPF to develop a variety of strategies to overcome or circumvent host defenses. While the interaction mechanisms between EPF and insects is well established, recent findings underscore that their interplay is more intricate than previously thought, especially evident across different stages of EPF infection. This review primarily focuses on the interplay between EPF and the insect defense strategies, centered around three infection stages: (1) Early infection stage: involving the pre-contact detection and avoidance behavior of EPF in insects, along with the induction of behavioral responses upon contact with the host cuticle; (2) Penetration and intra-hemolymph growth stage: involving the initiation of intricate cellular and humoral immune functions in insects, while symbiotic microbes can further contribute to host resistance; (3) Host insect's death stage: involving the ultimate confrontation between pathogens and insects. Infected insects strive to separate themselves from the healthy population, while pathogens rely on the infected insects to spread to new hosts. Also, we discuss a novel pest management strategy underlying the cooperation between EPF infection and disturbing the insect immune system. By enhancing our understanding of the intricate interplay between EPF and the insect, this review provides novel perspectives for EPF-mediated pest management and developing effective fungal insecticides.
Collapse
Affiliation(s)
- Meiqi Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jing Luo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Chong Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States
| | - Wei Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering (Ministry of Education), Guizhou University, Guiyang, China
| | - Letian Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
16
|
Zhou SO, Arunkumar R, Irfan A, Ding SD, Leitão AB, Jiggins FM. The evolution of constitutively active humoral immune defenses in Drosophila populations under high parasite pressure. PLoS Pathog 2024; 20:e1011729. [PMID: 38206983 PMCID: PMC10807768 DOI: 10.1371/journal.ppat.1011729] [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: 10/02/2023] [Revised: 01/24/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Both constitutive and inducible immune mechanisms are employed by hosts for defense against infection. Constitutive immunity allows for a faster response, but it comes with an associated cost that is always present. This trade-off between speed and fitness costs leads to the theoretical prediction that constitutive immunity will be favored where parasite exposure is frequent. We selected populations of Drosophila melanogaster under high parasite pressure from the parasitoid wasp Leptopilina boulardi. With RNA sequencing, we found the evolution of resistance in these populations was associated with them developing constitutively active humoral immunity, mediated by the larval fat body. Furthermore, these evolved populations were also able to induce gene expression in response to infection to a greater level, which indicates an overall more activated humoral immune response to parasitization. The anti-parasitoid immune response also relies on the JAK/STAT signaling pathway being activated in muscles following infection, and this induced response was only seen in populations that had evolved under high parasite pressure. We found that the cytokine Upd3, which induces this JAK/STAT response, is being expressed by immature lamellocytes. Furthermore, these immune cells became constitutively present when populations evolved resistance, potentially explaining why they gained the ability to activate JAK/STAT signaling. Thus, under intense parasitism, populations evolved resistance by increasing both constitutive and induced immune defenses, and there is likely an interplay between these two forms of immunity.
Collapse
Affiliation(s)
- Shuyu Olivia Zhou
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Ramesh Arunkumar
- Section of population genetics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Amina Irfan
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | | - Alexandre B. Leitão
- Champalimaud Foundation, Champalimaud Centre of the Unknown, Lisbon, Portugal
| | - Francis M. Jiggins
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Hwang SH, Jang HA, Kojour MAM, Yun K, Lee YS, Han YS, Jo YH. Effects of TmTak1 silencing on AMP production as an Imd pathway component in Tenebrio molitor. Sci Rep 2023; 13:18914. [PMID: 37919359 PMCID: PMC10622451 DOI: 10.1038/s41598-023-45978-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Mealworms beetles, Tenebrio molitor, are the limelight next-generation food for humans due to their high nutrient contents. Since Tenebrio molitor is used as feed for pets and livestock in addition to their ability to decompose polystyrene and plastic waste, it is recognized as an insect with an industrial core value. Therefore, it is important to study the immune mechanism related to the development and infection of mealworms for mass breeding purposes. The immune deficiency (Imd) signaling is one of the main pathways with pivotal roles in the production of antimicrobial peptides (AMPs). Transforming growth factor-β activated kinase (TAK1) is one of the Imd pathway components, forms a complex with TAK1 binding protein 2 (TAB2) to ultimately help activate the transcription factor Relish and eventually induce host to produce AMPs. Relatively, little has been revealed about TAK1 in insect models, especially in the T. molitor. Therefore, this study was conducted to elucidate the function of TmTak1 in T. molitor. Our results showed that the highest and lowest mRNA expression of TmTak1 were found in egg and young larvae respectively. The tissue-specific expression patterns were reported in the gut of T. molitor larvae and the fat bodies of adults. Systemic microbial challenge illustrated TmTak1 high expression following the fungal infection in all dissected tissues except for the whole body. However, silencing TmTak1 experiments showed that the survivability of T. molitor larvae affected significantly following Escherichia coli infection. Accordingly, AMP induction after TmTak1 knock down was mainly reported in the integument and the fat bodies.
Collapse
Affiliation(s)
- Su Hyeon Hwang
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Ho Am Jang
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, Chungnam, Republic of Korea
- Korea Native Animal Resources Utilization Convergence Research Institute (KNAR), Soonchunhyang University, Asan, Chungnam, Republic of Korea
| | - Maryam Ali Mohammadie Kojour
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Keunho Yun
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Seok Lee
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, Chungnam, Republic of Korea
- Korea Native Animal Resources Utilization Convergence Research Institute (KNAR), Soonchunhyang University, Asan, Chungnam, Republic of Korea
| | - Yeon Soo Han
- Division of Plant Biotechnology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Hun Jo
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, Chungnam, Republic of Korea.
- Korea Native Animal Resources Utilization Convergence Research Institute (KNAR), Soonchunhyang University, Asan, Chungnam, Republic of Korea.
| |
Collapse
|
18
|
Li S, Wang J, Tian X, Toufeeq S, Huang W. Immunometabolic regulation during the presence of microorganisms and parasitoids in insects. Front Immunol 2023; 14:905467. [PMID: 37818375 PMCID: PMC10560992 DOI: 10.3389/fimmu.2023.905467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Multicellular organisms live in environments containing diverse nutrients and a wide variety of microbial communities. On the one hand, the immune response of organisms can protect from the intrusion of exogenous microorganisms. On the other hand, the dynamic coordination of anabolism and catabolism of organisms is a necessary factor for growth and reproduction. Since the production of an immune response is an energy-intensive process, the activation of immune cells is accompanied by metabolic transformations that enable the rapid production of ATP and new biomolecules. In insects, the coordination of immunity and metabolism is the basis for insects to cope with environmental challenges and ensure normal growth, development and reproduction. During the activation of insect immune tissues by pathogenic microorganisms, not only the utilization of organic resources can be enhanced, but also the activated immune cells can usurp the nutrients of non-immune tissues by generating signals. At the same time, insects also have symbiotic bacteria in their body, which can affect insect physiology through immune-metabolic regulation. This paper reviews the research progress of insect immune-metabolism regulation from the perspective of insect tissues, such as fat body, gut and hemocytes. The effects of microorganisms (pathogenic bacteria/non-pathogenic bacteria) and parasitoids on immune-metabolism were elaborated here, which provide guidance to uncover immunometabolism mechanisms in insects and mammals. This work also provides insights to utilize immune-metabolism for the formulation of pest control strategies.
Collapse
Affiliation(s)
- Shirong Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- College of Life Sciences, Yan’an University, Yan’an, Shaanxi, China
| | - Jing Wang
- College of Life Sciences, Shangrao Normal University, Shangrao, China
| | - Xing Tian
- College of Life Sciences, Yan’an University, Yan’an, Shaanxi, China
| | - Shahzad Toufeeq
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Wuren Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
19
|
Ramelow J, Keleta Y, Niu G, Wang X, Li J. Plasmodium parasitophorous vacuole membrane protein Pfs16 promotes malaria transmission by silencing mosquito immunity. J Biol Chem 2023:104824. [PMID: 37196765 DOI: 10.1016/j.jbc.2023.104824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/07/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
With rising cases for the first time in years, malaria remains a significant public health burden. The sexual stage of the malaria parasite infects mosquitoes to transmit malaria from host to host. Hence, an infected mosquito plays an essential role in malaria transmission. Plasmodium falciparum is the most dominant and dangerous malaria pathogen. Previous studies identified a sexual stage-specific protein 16 (Pfs16) localized to the parasitophorous vacuole membrane (PVM). Here we elucidate the function of Pfs16 during malaria transmission. Our structural analysis identified Pfs16 as an alpha-helical integral membrane protein with one transmembrane domain connecting to two regions across PVM. ELISA assays showed that insect cell-expressed recombinant Pfs16 (rPfs16) interacted with An. gambiae midguts, and microscopy found that rPfs16 bound to midgut epithelial cells. Transmission-blocking assays demonstrated that polyclonal antibodies against Pfs16 significantly reduced the number of oocysts in mosquito midguts. However, on the contrary, feeding rPfs16 increased the number of oocysts. Further analysis revealed that Pfs16 reduced the activity of mosquito midgut caspase 3/7, a key enzyme in the mosquito Jun-N-terminal kinase (JNK) immune pathway. We conclude that Pfs16 facilitates parasites to invade mosquito midguts by actively silencing the mosquito's innate immunity through its interaction with the midgut epithelial cells. Therefore, Pfs16 is a potential target to control malaria transmission.
Collapse
Affiliation(s)
- Julian Ramelow
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Guodong Niu
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Xiaohong Wang
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Jun Li
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; Department of Biological Sciences, Florida International University, Miami, FL 33199, USA; Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
20
|
Salazar AM, Aparicio R, Clark RI, Rera M, Walker DW. Intestinal barrier dysfunction: an evolutionarily conserved hallmark of aging. Dis Model Mech 2023; 16:dmm049969. [PMID: 37144684 PMCID: PMC10184675 DOI: 10.1242/dmm.049969] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
A major challenge in the biology of aging is to understand how specific age-onset pathologies relate to the overall health of the organism. The integrity of the intestinal epithelium is essential for the wellbeing of the organism throughout life. In recent years, intestinal barrier dysfunction has emerged as an evolutionarily conserved feature of aged organisms, as reported in worms, flies, fish, rodents and primates. Moreover, age-onset intestinal barrier dysfunction has been linked to microbial alterations, elevated immune responses, metabolic alterations, systemic health decline and mortality. Here, we provide an overview of these findings. We discuss early work in the Drosophila model that sets the stage for examining the relationship between intestinal barrier integrity and systemic aging, then delve into research in other organisms. An emerging concept, supported by studies in both Drosophila and mice, is that directly targeting intestinal barrier integrity is sufficient to promote longevity. A better understanding of the causes and consequences of age-onset intestinal barrier dysfunction has significant relevance to the development of interventions to promote healthy aging.
Collapse
Affiliation(s)
- Anna M. Salazar
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA 23606, USA
| | - Ricardo Aparicio
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Rebecca I. Clark
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Michael Rera
- Université de Paris, Inserm U1284, Center for Research and Interdisciplinarity, Paris 75004, France
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Bland ML. Regulating metabolism to shape immune function: Lessons from Drosophila. Semin Cell Dev Biol 2023; 138:128-141. [PMID: 35440411 PMCID: PMC10617008 DOI: 10.1016/j.semcdb.2022.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/21/2022] [Accepted: 04/03/2022] [Indexed: 12/14/2022]
Abstract
Infection with pathogenic microbes is a severe threat that hosts manage by activating the innate immune response. In Drosophila melanogaster, the Toll and Imd signaling pathways are activated by pathogen-associated molecular patterns to initiate cellular and humoral immune processes that neutralize and kill invaders. The Toll and Imd signaling pathways operate in organs such as fat body and gut that control host nutrient metabolism, and infections or genetic activation of Toll and Imd signaling also induce wide-ranging changes in host lipid, carbohydrate and protein metabolism. Metabolic regulation by immune signaling can confer resistance to or tolerance of infection, but it can also lead to pathology and susceptibility to infection. These immunometabolic phenotypes are described in this review, as are changes in endocrine signaling and gene regulation that mediate survival during infection. Future work in the field is anticipated to determine key variables such as sex, dietary nutrients, life stage, and pathogen characteristics that modify immunometabolic phenotypes and, importantly, to uncover the mechanisms used by the immune system to regulate metabolism.
Collapse
Affiliation(s)
- Michelle L Bland
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, United States.
| |
Collapse
|
22
|
Liu Y, Li X, Lin L. Transcriptome of the pygmy grasshopper Formosatettix qinlingensis (Orthoptera: Tetrigidae). PeerJ 2023; 11:e15123. [PMID: 37016680 PMCID: PMC10066883 DOI: 10.7717/peerj.15123] [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: 07/27/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Formosatettix qinlingensis (Zheng, 1982) is a tiny grasshopper endemic to Qinling in China. For further study of its transcriptomic features, we obtained RNA-Seq data by Illumina HiSeq X Ten sequencing platform. Firstly, transcriptomic analysis showed that transcriptome read numbers of two female and one male samples were 25,043,314, 24,429,905, and 25,034,457, respectively. We assembled 65,977 unigenes, their average length was 1,072.09 bp, and the length of N50 was 2,031 bp. The average lengths of F. qinlingensis female and male unigenes were 911.30 bp, and 941.82 bp, and the N50 lengths were 1,745 bp and 1,735 bp, respectively. Eight databases were used to annotate the functions of unigenes, and 23,268 functional unigenes were obtained. Besides, we also studied the body color, immunity and insecticide resistance of F. qinlingensis. Thirty-nine pigment-related genes were annotated. Some immunity genes and signaling pathways were found, such as JAK-STAT and Toll-LIKE receptor signaling pathways. There are also some insecticide resistance genes and signal pathways, like nAChR, GST and DDT. Further, some of these genes were differentially expressed in female and male samples, including pigment, immunity and insecticide resistance. The transcriptomic study of F. qinlingensis will provide data reference for gene prediction and molecular expression study of other Tetrigidae species in the future. Differential genetic screening of males and females provides a basis for studying sex and immune balance in insects.
Collapse
Affiliation(s)
- Yuxin Liu
- Shaanxi Normal University, Xi’an, China
| | | | | |
Collapse
|
23
|
Wijerathna HMSM, Nadarajapillai K, Shanaka KASN, Kasthuriarachchi TDW, Jung S, Lee S, Lee J. Molecular characterization and immune response of suppressor of cytokine signaling 5b from redlip mullet (Planiliza haematocheilus): Disclosing its anti-viral potential and effect on cell proliferation. FISH & SHELLFISH IMMUNOLOGY 2023; 134:108629. [PMID: 36822381 DOI: 10.1016/j.fsi.2023.108629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/27/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
The suppressor of cytokine signaling (SOCS) proteins family comprising eight proteins (SOCS1-7 and cytokine-inducible SH2-containing (CIS)) are classical feedback inhibitors of cytokine signaling. Although the biological role of CIS and SOCS1-3 have been extensively studied, the biological functions of SOCS4-7 remain unclear. Here, we elucidated the molecular characteristics, expression profile, immune response, anti-viral potential, and effect on cell proliferation of Phsocs5b, a member of the SOCS protein family from redlip mullet (Planiliza haematocheilus); phsocs5b comprised 1695 nucleotides. It was 564 amino acids long with a molecular weight of 62.3 kDa and a theoretical isoelectric point of 8.95. Like SOCS4-7 proteins, Phsocs5b comprised an SH2 domain, SOCS box domain, and a long N-terminal. SH2 domain is highly identical to its orthologs in other vertebrates. Phsocs5b, highly expressed in the brain tissue, was localized in the cytoplasm. Temporal changes in phsocs5b expression were observed following immune stimulation with polyinosinic: polycytidylic acid, lipopolysaccharide, and Lactococcus garvieae. In FHM cells, Phsocs5b overexpression suppressed the viral hemorrhagic septicemia virus (VHSV) infection and epidermal growth factor receptor (egfr) expression but increased the mRNA levels of pi3k, akt, pro-inflammatory cytokines (il1β and il8), and anti-viral genes (isg15 and ifn). Overall, our findings suggest that Phsocs5b attenuates VHSV infection, either by hindering the cell entry via degradation of Egfr, enhancing pro-inflammatory cytokines and anti-viral factor production, or both. The results also indicated that Phsocs5b could directly activate Pi3k/Akt pathway by itself, thus enhancing the proliferation and migration of cells. Taken together, Phsocs5b may be considered a potential therapeutic target to enhance immune responses while positively regulating the proliferation and migration of cells.
Collapse
Affiliation(s)
- H M S M Wijerathna
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Kishanthini Nadarajapillai
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - K A S N Shanaka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - T D W Kasthuriarachchi
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Seongdo Lee
- National Fishery Product Quality Management Service, Busan, 49111, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
24
|
Jatuyosporn T, Laohawutthichai P, Romo JPO, Gallardo-Becerra L, Lopez FS, Tassanakajon A, Ochoa-Leyva A, Krusong K. White spot syndrome virus impact on the expression of immune genes and gut microbiome of black tiger shrimp Penaeus monodon. Sci Rep 2023; 13:996. [PMID: 36653369 PMCID: PMC9849358 DOI: 10.1038/s41598-023-27906-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
The gut microbiome plays an essential role in the immune system of invertebrates and vertebrates. Pre and pro-biotics could enhance the shrimp immune system by increasing the phenoloxidase (PO), prophenoloxidase (ProPO), and superoxide dismutase activities. During viral infection, the host immune system alteration could influence the gut microbiome composition and probably lead to other pathogenic infections. Since the JAK/STAT pathway is involved in white spot syndrome virus (WSSV) infection, we investigated the intestine immune genes of STAT-silenced shrimp. During WSSV infection, expression levels of PmVago1, PmDoral, and PmSpätzle in PmSTAT-silenced shrimp were higher than normal. In addition, the transcription levels of antimicrobial peptides, including crustinPm1, crustinPm7, and PmPEN3, were higher in WSSV-challenged PmSTAT-silenced shrimp than the WSSV-infected normal shrimp. Meanwhile, PmSTAT silencing suppressed PmProPO1, PmProPO2, and PmPPAE1 expressions during WSSV infection. The microbiota from four shrimp tested groups (control group, WSSV-infected, PmSTAT-silenced, and PmSTAT-silenced infected by WSSV) was significantly different, with decreasing richness and diversity due to WSSV infection. The relative abundance of Bacteroidetes, Actinobacteria, and Planctomycetes was reduced in WSSV-challenged shrimp. However, at the species level, P. damselae, a pathogen to human and marine animals, significantly increased in WSSV-challenged shrimp. In constrast, Shewanella algae, a shrimp probiotic, was decreased in WSSV groups. In addition, the microbiota structure between control and PmSTAT-silenced shrimp was significantly different, suggesting the importance of STAT to maintain the homeostasis interaction with the microbiota.
Collapse
Affiliation(s)
- Thapanan Jatuyosporn
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.,Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pasunee Laohawutthichai
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.,Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Juan Pablo Ochoa Romo
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad #2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, Mexico
| | - Luigui Gallardo-Becerra
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad #2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, Mexico
| | - Filiberto Sánchez Lopez
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad #2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, Mexico
| | - Anchalee Tassanakajon
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Adrian Ochoa-Leyva
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad #2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, Mexico.
| | - Kuakarun Krusong
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
25
|
Ding SD, Leitão AB, Day JP, Arunkumar R, Phillips M, Zhou SO, Jiggins FM. Trans-regulatory changes underpin the evolution of the Drosophila immune response. PLoS Genet 2022; 18:e1010453. [PMID: 36342922 PMCID: PMC9671443 DOI: 10.1371/journal.pgen.1010453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/17/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022] Open
Abstract
When an animal is infected, the expression of a large suite of genes is changed, resulting in an immune response that can defend the host. Despite much evidence that the sequence of proteins in the immune system can evolve rapidly, the evolution of gene expression is comparatively poorly understood. We therefore investigated the transcriptional response to parasitoid wasp infection in Drosophila simulans and D. sechellia. Although these species are closely related, there has been a large scale divergence in the expression of immune-responsive genes in their two main immune tissues, the fat body and hemocytes. Many genes, including those encoding molecules that directly kill pathogens, have cis regulatory changes, frequently resulting in large differences in their expression in the two species. However, these changes in cis regulation overwhelmingly affected gene expression in immune-challenged and uninfected animals alike. Divergence in the response to infection was controlled in trans. We argue that altering trans-regulatory factors, such as signalling pathways or immune modulators, may allow natural selection to alter the expression of large numbers of immune-responsive genes in a coordinated fashion.
Collapse
Affiliation(s)
| | - Alexandre B. Leitão
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Champalimaud Foundation, Lisbon, Portugal
| | - Jonathan P. Day
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Ramesh Arunkumar
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Morgan Phillips
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Shuyu Olivia Zhou
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Francis M. Jiggins
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
26
|
Arch M, Vidal M, Koiffman R, Melkie ST, Cardona PJ. Drosophila melanogaster as a model to study innate immune memory. Front Microbiol 2022; 13:991678. [PMID: 36338030 PMCID: PMC9630750 DOI: 10.3389/fmicb.2022.991678] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/03/2022] [Indexed: 09/12/2023] Open
Abstract
Over the last decades, research regarding innate immune responses has gained increasing importance. A growing body of evidence supports the notion that the innate arm of the immune system could show memory traits. Such traits are thought to be conserved throughout evolution and provide a survival advantage. Several models are available to study these mechanisms. Among them, we find the fruit fly, Drosophila melanogaster. This non-mammalian model has been widely used for innate immune research since it naturally lacks an adaptive response. Here, we aim to review the latest advances in the study of the memory mechanisms of the innate immune response using this animal model.
Collapse
Affiliation(s)
- Marta Arch
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Maria Vidal
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Romina Koiffman
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- UCBL, UnivLyon, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Solomon Tibebu Melkie
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- UCBL, UnivLyon, Université Claude Bernard Lyon 1 (UCBL1), Villeurbanne, France
| | - Pere-Joan Cardona
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
27
|
Characterization of New Defensin Antimicrobial Peptides and Their Expression in Bed Bugs in Response to Bacterial Ingestion and Injection. Int J Mol Sci 2022; 23:ijms231911505. [PMID: 36232802 PMCID: PMC9570333 DOI: 10.3390/ijms231911505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Common bed bugs, Cimex lectularius, can carry, but do not transmit, pathogens to the vertebrate hosts on which they feed. Some components of the innate immune system of bed bugs, such as antimicrobial peptides (AMPs), eliminate the pathogens. Here, we determined the molecular characteristics, structural properties, and phylogenetic relatedness of two new defensins (CL-defensin1 (XP_024085718.1), CL-defensin2 (XP_014240919.1)), and two new defensin isoforms (CL-defensin3a (XP_014240918.1), CL-defensin3b (XP_024083729.1)). The complete amino acid sequences of CL-defensin1, CL-defensin2, CL-defensin3a, and CL-defensin3b are strongly conserved, with only minor differences in their signal and pro-peptide regions. We used a combination of comparative transcriptomics and real-time quantitative PCR to evaluate the expression of these defensins in the midguts and the rest of the body of insects that had been injected with bacteria or had ingested blood containing the Gram-positive (Gr+) bacterium Bacillus subtilis and the Gram-negative (Gr–) bacterium Escherichia coli. We demonstrate, for the first time, sex-specific and immunization mode-specific upregulation of bed bug defensins in response to injection or ingestion of Gr+ or Gr– bacteria. Understanding the components, such as these defensins, of the bed bugs’ innate immune systems in response to pathogens may help unravel why bed bugs do not transmit pathogens to vertebrates.
Collapse
|
28
|
Zhang C, Jin Y, Marchetti M, Lewis MR, Hammouda OT, Edgar BA. EGFR signaling activates intestinal stem cells by promoting mitochondrial biogenesis and β-oxidation. Curr Biol 2022; 32:3704-3719.e7. [PMID: 35896119 PMCID: PMC10117080 DOI: 10.1016/j.cub.2022.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 05/11/2022] [Accepted: 07/04/2022] [Indexed: 10/16/2022]
Abstract
EGFR-RAS-ERK signaling promotes growth and proliferation in many cell types, and genetic hyperactivation of RAS-ERK signaling drives many cancers. Yet, despite intensive study of upstream components in EGFR signal transduction, the identities and functions of downstream effectors in the pathway are poorly understood. In Drosophila intestinal stem cells (ISCs), the transcriptional repressor Capicua (Cic) and its targets, the ETS-type transcriptional activators Pointed (pnt) and Ets21C, are essential downstream effectors of mitogenic EGFR signaling. Here, we show that these factors promote EGFR-dependent metabolic changes that increase ISC mass, mitochondrial growth, and mitochondrial activity. Gene target analysis using RNA and DamID sequencing revealed that Pnt and Ets21C directly upregulate not only DNA replication and cell cycle genes but also genes for oxidative phosphorylation, the TCA cycle, and fatty acid beta-oxidation. Metabolite analysis substantiated these metabolic functions. The mitochondrial transcription factor B2 (mtTFB2), a direct target of Pnt, was required and partially sufficient for EGFR-driven ISC growth, mitochondrial biogenesis, and proliferation. MEK-dependent EGF signaling stimulated mitochondrial biogenesis in human RPE-1 cells, indicating the conservation of these metabolic effects. This work illustrates how EGFR signaling alters metabolism to coordinately activate cell growth and cell division.
Collapse
Affiliation(s)
- Chenge Zhang
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Center for Molecular Biology, Heidelberg University (ZMBH) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Yinhua Jin
- Center for Molecular Biology, Heidelberg University (ZMBH) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Department of Developmental Biology, Howard Hughes Medical Institute, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marco Marchetti
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Center for Molecular Biology, Heidelberg University (ZMBH) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mitchell R Lewis
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Omar T Hammouda
- Center for Molecular Biology, Heidelberg University (ZMBH) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Centre for Organismal Studies Heidelberg & Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Bruce A Edgar
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA; Center for Molecular Biology, Heidelberg University (ZMBH) & German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
29
|
Aalto A, Martínez‐Chacón G, Kietz C, Tsyganova N, Kreutzer J, Kallio P, Broemer M, Meinander A. M1-linked ubiquitination facilitates NF-κB activation and survival during sterile inflammation. FEBS J 2022; 289:5180-5197. [PMID: 35263507 PMCID: PMC9543601 DOI: 10.1111/febs.16425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 01/03/2023]
Abstract
Methionine 1 (M1)-linked ubiquitination plays a key role in the regulation of inflammatory nuclear factor-κB (NF-κB) signalling and is important for clearance of pathogen infection in Drosophila melanogaster. M1-linked ubiquitin (M1-Ub) chains are assembled by the linear ubiquitin E3 ligase (LUBEL) in flies. Here, we have studied the role of LUBEL in sterile inflammation induced by different types of cellular stresses. We have found that the LUBEL catalyses formation of M1-Ub chains in response to hypoxic, oxidative and mechanical stress conditions. LUBEL is shown to be important for flies to survive low oxygen conditions and paraquat-induced oxidative stress. This protective action seems to be driven by stress-induced activation of the NF-κB transcription factor Relish via the immune deficiency (Imd) pathway. In addition to LUBEL, the intracellular mediators of Relish activation, including the transforming growth factor activating kinase 1 (Tak1), Drosophila inhibitor of apoptosis (IAP) Diap2, the IκB kinase γ (IKKγ) Kenny and the initiator caspase Death-related ced-3/Nedd2-like protein (Dredd), but not the membrane receptor peptidoglycan recognition protein (PGRP)-LC, are shown to be required for sterile inflammatory response and survival. Finally, we showed that the stress-induced upregulation of M1-Ub chains in response to hypoxia, oxidative and mechanical stress is also induced in mammalian cells and protects from stress-induced cell death. Taken together, our results suggest that M1-Ub chains are important for NF-κB signalling in inflammation induced by stress conditions often observed in chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Anna Aalto
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | | | - Christa Kietz
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | - Nadezhda Tsyganova
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
| | - Joose Kreutzer
- Faculty of Medicine and Health TechnologyBioMediTechTampere UniversityFinland
| | - Pasi Kallio
- Faculty of Medicine and Health TechnologyBioMediTechTampere UniversityFinland
| | - Meike Broemer
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Annika Meinander
- Faculty of Science and Engineering, Cell BiologyÅbo Akademi University, BioCityTurkuFinland
- InFLAMES Research Flagship CenterÅbo Akademi UniversityTurkuFinland
| |
Collapse
|
30
|
Yan Y, Sigle LT, Rinker DC, Estévez-Lao TY, Capra JA, Hillyer JF. The immune deficiency and c-Jun N-terminal kinase pathways drive the functional integration of the immune and circulatory systems of mosquitoes. Open Biol 2022; 12:220111. [PMID: 36069078 PMCID: PMC9449813 DOI: 10.1098/rsob.220111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune and circulatory systems of animals are functionally integrated. In mammals, the spleen and lymph nodes filter and destroy microbes circulating in the blood and lymph, respectively. In insects, immune cells that surround the heart valves (ostia), called periostial haemocytes, destroy pathogens in the areas of the body that experience the swiftest haemolymph (blood) flow. An infection recruits additional periostial haemocytes, amplifying heart-associated immune responses. Although the structural mechanics of periostial haemocyte aggregation have been defined, the genetic factors that regulate this process remain less understood. Here, we conducted RNA sequencing in the African malaria mosquito, Anopheles gambiae, and discovered that an infection upregulates multiple components of the immune deficiency (IMD) and c-Jun N-terminal kinase (JNK) pathways in the heart with periostial haemocytes. This upregulation is greater in the heart with periostial haemocytes than in the circulating haemocytes or the entire abdomen. RNA interference-based knockdown then showed that the IMD and JNK pathways drive periostial haemocyte aggregation and alter phagocytosis and melanization on the heart, thereby demonstrating that these pathways regulate the functional integration between the immune and circulatory systems. Understanding how insects fight infection lays the foundation for novel strategies that could protect beneficial insects and harm detrimental ones.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Leah T. Sigle
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - David C. Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - John A. Capra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA,Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
31
|
Li T, Li H, Wu Y, Li S, Yuan G, Xu P. Identification of a Novel Densovirus in Aphid, and Uncovering the Possible Antiviral Process During Its Infection. Front Immunol 2022; 13:905628. [PMID: 35757766 PMCID: PMC9218065 DOI: 10.3389/fimmu.2022.905628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Densoviruses (DVs) are single-stranded DNA viruses and exclusively happen in invertebrates. Most of DVs reported in insects are pathogenic to their native hosts, however, no pathogenic effect of them has been examined in vertebrates. Hence, DVs are the potential agents used in pest managements. Aphids are the primary vectors of plant viruses. In this study, we identified a novel DV in Chinese Sitobion miscanthi population, provisionally named “Sitobion miscanthi densovirus” (SmDV). Taxonomically, SmDV belongs to genus Hemiambidensovirus. In S. miscanthi, SmDV is hosted in diverse cells and can be horizontally transmitted via wheat feeding. Subject to SmDV, aphids activate their intrinsic antiviral autophagy pathway. Grouped with ascorbate and aldarate metabolism, chlorophyll metabolism, p450 related drug metabolism, and retinoid metabolism, aphids form a complex immune network response to the infection of SmDV. Obviously, it works as elder aphids still alive even they contain the highest examined concentration of SmDV. This study provides a foundation for the identifications of novel DVs, and further improves the understanding of the molecular interactions between insects and DVs.
Collapse
Affiliation(s)
- Tong Li
- Institute of Plant Protection, Henan Key Laboratory of Crop Pest Control, Key Laboratory of Integrated Pest Management on Crops in Southern Region of North China, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Haichao Li
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China.,Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Yuqing Wu
- Institute of Plant Protection, Henan Key Laboratory of Crop Pest Control, Key Laboratory of Integrated Pest Management on Crops in Southern Region of North China, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Shaojian Li
- Institute of Plant Protection, Henan Key Laboratory of Crop Pest Control, Key Laboratory of Integrated Pest Management on Crops in Southern Region of North China, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Guohui Yuan
- College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Pengjun Xu
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, China
| |
Collapse
|
32
|
Waring AL, Hill J, Allen BM, Bretz NM, Le N, Kr P, Fuss D, Mortimer NT. Meta-Analysis of Immune Induced Gene Expression Changes in Diverse Drosophila melanogaster Innate Immune Responses. INSECTS 2022; 13:insects13050490. [PMID: 35621824 PMCID: PMC9147463 DOI: 10.3390/insects13050490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/05/2022]
Abstract
Simple Summary Organisms can be infected by a wide range of pathogens, including bacteria, viruses, and parasites. Following infection, the host mounts an immune response to attempt to eliminate the pathogen. These responses are often specific to the type of pathogen and mediated by the expression of specialized genes. We have characterized the expression changes induced in host Drosophila fruit flies following infection by multiple types of pathogens, and identified a small number of genes that show expression changes in each infection. This includes genes that are known to be involved in pathogen resistance, and others that have not been previously studied as immune response genes. These findings provide new insight into transcriptional changes that accompany Drosophila immunity. They may suggest possible roles for the differentially expressed genes in innate immune responses to diverse classes of pathogens, and serve to identify candidate genes for further empirical study of these processes. Abstract Organisms are commonly infected by a diverse array of pathogens and mount functionally distinct responses to each of these varied immune challenges. Host immune responses are characterized by the induction of gene expression, however, the extent to which expression changes are shared among responses to distinct pathogens is largely unknown. To examine this, we performed meta-analysis of gene expression data collected from Drosophila melanogaster following infection with a wide array of pathogens. We identified 62 genes that are significantly induced by infection. While many of these infection-induced genes encode known immune response factors, we also identified 21 genes that have not been previously associated with host immunity. Examination of the upstream flanking sequences of the infection-induced genes lead to the identification of two conserved enhancer sites. These sites correspond to conserved binding sites for GATA and nuclear factor κB (NFκB) family transcription factors and are associated with higher levels of transcript induction. We further identified 31 genes with predicted functions in metabolism and organismal development that are significantly downregulated following infection by diverse pathogens. Our study identifies conserved gene expression changes in Drosophila melanogaster following infection with varied pathogens, and transcription factor families that may regulate this immune induction.
Collapse
|
33
|
Abstract
Inflammatory response in Drosophila to sterile (axenic) injury in embryos and adults has received some attention in recent years, and most concentrate on the events at the injury site. Here we focus on the effect sterile injury has on the hematopoietic organ, the lymph gland, and the circulating blood cells in the larva, the developmental stage at which major events of hematopoiesis are evident. In mammals, injury activates Toll-like receptor/NF-κB signaling in macrophages, which then express and secrete secondary, proinflammatory cytokines. In Drosophila larvae, distal puncture injury of the body wall epidermis causes a rapid activation of Toll and Jun kinase (JNK) signaling throughout the hematopoietic system and the differentiation of a unique blood cell type, the lamellocyte. Furthermore, we find that Toll and JNK signaling are coupled in their activation. Secondary to this Toll/JNK response, a cytokine, Upd3, is induced as a Toll pathway transcriptional target, which then promotes JAK/STAT signaling within the blood cells. Toll and JAK/STAT signaling are required for the emergence of the injury-induced lamellocytes. This is akin to the derivation of specialized macrophages in mammalian systems. Upstream, at the injury site, a Duox- and peroxide-dependent signal causes the activation of the proteases Grass and SPE, needed for the activation of the Toll-ligand Spz, but microbial sensors or the proteases most closely associated with them during septic injury are not involved in the axenic inflammatory response.
Collapse
|
34
|
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
|
35
|
Chen Z, Zhang W, Wang F, Mu R, Wen D. Sestrin protects Drosophila midgut from mercury chloride-induced damage by inhibiting oxidative stress and stimulating intestinal regeneration. Comp Biochem Physiol C Toxicol Pharmacol 2021; 248:109083. [PMID: 34089877 DOI: 10.1016/j.cbpc.2021.109083] [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: 02/09/2021] [Revised: 04/27/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
Overproduction of the deleterious reactive oxygen species (ROS) is one of the major causes of mercury, a heavy metal with diverse applications and environmental presence, induced neuronal and gastrointestinal adversities in exposed organism including Drosophila melanogaster. Sestrin, an oxidative stress responsive gene, emerges as a novel player in the management of oxidative stress response. Due to limited information regarding the role of sestrin in mercury-induced gastrointestinal adversities, it was hypothesized that modulation of sestrin may improve the mercury-induced gastrointestinal adversities in Drosophila. Here, we fed Drosophila with 400 μM HgCl2 and found that sestrin transcriptional level was significantly increased in midguts. Sestrin knockdown in HgCl2-exposed midguts decreased survival rates and climbing ability of flies, and inhibited superoxide dismutase and glutathione-S-transferase activities of midgut epithelieum. Meanwhile, sestrin knockdown in midgut aggravated the HgCl2-induced disruption of intestinal organization by worsening ROS production and cell apoptosis. Immunohistochemical staining data revealed that sestrin knockdown inhibited intestinal stem cell division in HgCl2-exposed midguts. Furthermore, JNK signaling was found to mediated sestrin expression in midgut. Taken together, the study demonstrated that sestrin protects Drosophila midgut from HgCl2-induced oxidative damage by inhibiting ROS production and stimulating the tissue regeneration program under regulation of JNK signaling pathway. This work suggests therapeutic implications of sestrin against heavy metal-induced gastrointestinal adversities in mammals.
Collapse
Affiliation(s)
- Zhi Chen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| | - Wen Zhang
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China
| | - Fen Wang
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China
| | - Ren Mu
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| | - Di Wen
- College of Biological Science and Agriculture, Qiannan Normal University for Nationalities, Duyun 558000, China.
| |
Collapse
|
36
|
Ramesh P, Dey NS, Kanwal A, Mandal S, Mandal L. Relish plays a dynamic role in the niche to modulate Drosophila blood progenitor homeostasis in development and infection. eLife 2021; 10:67158. [PMID: 34292149 PMCID: PMC8363268 DOI: 10.7554/elife.67158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Immune challenges demand the gearing up of basal hematopoiesis to combat infection. Little is known about how during development, this switch is achieved to take care of the insult. Here, we show that the hematopoietic niche of the larval lymph gland of Drosophila senses immune challenge and reacts to it quickly through the nuclear factor-κB (NF-κB), Relish, a component of the immune deficiency (Imd) pathway. During development, Relish is triggered by ecdysone signaling in the hematopoietic niche to maintain the blood progenitors. Loss of Relish causes an alteration in the cytoskeletal architecture of the niche cells in a Jun Kinase-dependent manner, resulting in the trapping of Hh implicated in progenitor maintenance. Notably, during infection, downregulation of Relish in the niche tilts the maintenance program toward precocious differentiation, thereby bolstering the cellular arm of the immune response.
Collapse
Affiliation(s)
- Parvathy Ramesh
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, India.,Developmental Genetics Laboratory, IISER Mohali, SAS Nagar, Punjab, India
| | - Nidhi Sharma Dey
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, India.,Developmental Genetics Laboratory, IISER Mohali, SAS Nagar, Punjab, India
| | - Aditya Kanwal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, India.,Developmental Genetics Laboratory, IISER Mohali, SAS Nagar, Punjab, India
| | - Sudip Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, India.,Molecular Cell and Developmental Biology Laboratory, IISER Mohali, SAS Nagar, Punjab, India
| | - Lolitika Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, Knowledge City, India.,Developmental Genetics Laboratory, IISER Mohali, SAS Nagar, Punjab, India
| |
Collapse
|
37
|
Microenvironmental innate immune signaling and cell mechanical responses promote tumor growth. Dev Cell 2021; 56:1884-1899.e5. [PMID: 34197724 DOI: 10.1016/j.devcel.2021.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/01/2021] [Accepted: 06/09/2021] [Indexed: 01/08/2023]
Abstract
Tissue homeostasis is achieved by balancing stem cell maintenance, cell proliferation and differentiation, as well as the purging of damaged cells. Elimination of unfit cells maintains tissue health; however, the underlying mechanisms driving competitive growth when homeostasis fails, for example, during tumorigenesis, remain largely unresolved. Here, using a Drosophila intestinal model, we find that tumor cells outcompete nearby enterocytes (ECs) by influencing cell adhesion and contractility. This process relies on activating the immune-responsive Relish/NF-κB pathway to induce EC delamination and requires a JNK-dependent transcriptional upregulation of the peptidoglycan recognition protein PGRP-LA. Consequently, in organisms with impaired PGRP-LA function, tumor growth is delayed and lifespan extended. Our study identifies a non-cell-autonomous role for a JNK/PGRP-LA/Relish signaling axis in mediating death of neighboring normal cells to facilitate tumor growth. We propose that intestinal tumors "hijack" innate immune signaling to eliminate enterocytes in order to support their own growth.
Collapse
|
38
|
Comparative transcriptome analysis reveals a potential mechanism for host nutritional manipulation after parasitization by Leptopilina boulardi. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 39:100862. [PMID: 34120097 DOI: 10.1016/j.cbd.2021.100862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/13/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023]
Abstract
Parasitoids have been extensively found to manipulate nutrient amounts of their hosts to benefit their own development and survival, but the underlying mechanisms are largely unknown. Leptopilina boulardi (Hymenoptera: Figitidae) is a larval-pupal endoparasitoid wasp of Drosophila melanogaster whose survival relies on the nutrients provided by its Drosophila host. Here, we used RNA-seq to compare the gene expression levels of the host midgut at 24 h and 48 h post L. boulardi parasitization. We obtained 95 and 191 differentially expressed genes (DEGs) in the parasitized host midgut at 24 h and 48 h post L. boulardi parasitization, respectively. A KEGG analysis revealed that several metabolic pathways were significantly enriched in the upregulated DEGs, and these pathways included "starch and sucrose metabolism" and "galactose metabolism". A functional annotation analysis showed that four classes of genes involved in carbohydrate digestion process had increased expression levels in the midgut post L.boulardi parasitization than nonparasitized groups: glucosidase, mannosidase, chitinase and amylase. Genes involved in protein digestion process were also found among the DEGs, and most of these genes, which belonged to the metallopeptidase and serine-type endopeptidase families, were found at higher expression levels in the parasitized host midgut comparing with nonparasitized hosts. Moreover, some immune genes, particularly those involved in the Toll and Imd pathways, also exhibited high expression levels after L.boulardi parasitization. Our study provides large-scale transcriptome data and identifies sets of DEGs between parasitized and nonparasitized host midgut tissues at 24 h and 48 h post L. boulardi parasitization. These resources help improve our understanding of how parasitoid infection affects the nutrient components in the hosts.
Collapse
|
39
|
Rosendo Machado S, van der Most T, Miesen P. Genetic determinants of antiviral immunity in dipteran insects - Compiling the experimental evidence. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104010. [PMID: 33476667 DOI: 10.1016/j.dci.2021.104010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
The genetic basis of antiviral immunity in dipteran insects is extensively studied in Drosophila melanogaster and advanced technologies for genetic manipulation allow a better characterization of immune responses also in non-model insect species. Especially, immunity in vector mosquitoes is recently in the spotlight, due to the medical impact that these insects have by transmitting viruses and other pathogens. Here, we review the current state of experimental evidence that supports antiviral functions for immune genes acting in different cellular pathways. We discuss the well-characterized RNA interference mechanism along with the less well-defined JAK-STAT, Toll, and IMD signaling pathways. Furthermore, we highlight the initial evidence for antiviral activity observed for the autophagy pathway, transcriptional pausing, as well as piRNA production from endogenous viral elements. We focus our review on studies from Drosophila and mosquito species from the lineages Aedes, Culex, and Anopheles, which contain major vector species responsible for virus transmission.
Collapse
Affiliation(s)
- Samara Rosendo Machado
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Tom van der Most
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands.
| |
Collapse
|
40
|
Ferguson M, Foley E. Microbial recognition regulates intestinal epithelial growth in homeostasis and disease. FEBS J 2021; 289:3666-3691. [PMID: 33977656 DOI: 10.1111/febs.15910] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/06/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
The intestine is constantly exposed to a dynamic community of microbes. Intestinal epithelial cells respond to microbes through evolutionarily conserved recognition pathways, such as the immune deficiency (IMD) pathway of Drosophila, the Toll-like receptor (TLR) response of flies and vertebrates, and the vertebrate nucleotide-binding oligomerization domain (NOD) pathway. Microbial recognition pathways are tightly controlled to respond effectively to pathogens, tolerate the microbiome, and limit intestinal disease. In this review, we focus on contributions of different model organisms to our understanding of how epithelial microbe recognition impacts intestinal proliferation and differentiation in homeostasis and disease. In particular, we compare how microbes and subsequent recognition by the intestine influences barrier integrity, intestinal repair and tumorigenesis in Drosophila, zebrafish, mice, and organoids. In addition, we discuss the importance of microbial recognition in homeostatic intestinal growth and discuss how immune pathways directly impact stem cell and crypt dynamics.
Collapse
Affiliation(s)
- Meghan Ferguson
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
41
|
Schlamp F, Delbare SYN, Early AM, Wells MT, Basu S, Clark AG. Dense time-course gene expression profiling of the Drosophila melanogaster innate immune response. BMC Genomics 2021; 22:304. [PMID: 33902461 PMCID: PMC8074482 DOI: 10.1186/s12864-021-07593-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Immune responses need to be initiated rapidly, and maintained as needed, to prevent establishment and growth of infections. At the same time, resources need to be balanced with other physiological processes. On the level of transcription, studies have shown that this balancing act is reflected in tight control of the initiation kinetics and shutdown dynamics of specific immune genes. RESULTS To investigate genome-wide expression dynamics and trade-offs after infection at a high temporal resolution, we performed an RNA-seq time course on D. melanogaster with 20 time points post Imd stimulation. A combination of methods, including spline fitting, cluster analysis, and Granger causality inference, allowed detailed dissection of expression profiles, lead-lag interactions, and functional annotation of genes through guilt-by-association. We identified Imd-responsive genes and co-expressed, less well characterized genes, with an immediate-early response and sustained up-regulation up to 5 days after stimulation. In contrast, stress response and Toll-responsive genes, among which were Bomanins, demonstrated early and transient responses. We further observed a strong trade-off with metabolic genes, which strikingly recovered to pre-infection levels before the immune response was fully resolved. CONCLUSIONS This high-dimensional dataset enabled the comprehensive study of immune response dynamics through the parallel application of multiple temporal data analysis methods. The well annotated data set should also serve as a useful resource for further investigation of the D. melanogaster innate immune response, and for the development of methods for analysis of a post-stress transcriptional response time-series at whole-genome scale.
Collapse
Affiliation(s)
- Florencia Schlamp
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| | | | - Angela M Early
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Martin T Wells
- Statistics and Data Science, Cornell University, Ithaca, NY, USA
| | - Sumanta Basu
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| | - Andrew G Clark
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
- Statistics and Data Science, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
42
|
Yan J, Zhao N, Yang Z, Li Y, Bai H, Zou W, Zhang K, Huang X. A trade-off switch of two immunological memories in Caenorhabditis elegans reinfected by bacterial pathogens. J Biol Chem 2020; 295:17323-17336. [PMID: 33051209 PMCID: PMC7863904 DOI: 10.1074/jbc.ra120.013923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/25/2020] [Indexed: 11/06/2022] Open
Abstract
Recent studies have suggested that innate immune responses exhibit characteristics associated with memory linked to modulations in both vertebrates and invertebrates. However, the diverse evolutionary paths taken, particularly within the invertebrate taxa, should lead to similarly diverse innate immunity memory processes. Our understanding of innate immune memory in invertebrates primarily comes from studies of the fruit fly Drosophila melanogaster, the generality of which is unclear. Caenorhabditis elegans typically inhabits soil harboring a variety of fatal microbial pathogens; for this invertebrate, the innate immune system and aversive behavior are the major defensive strategies against microbial infection. However, their characteristics of immunological memory remains infantile. Here we discovered an immunological memory that promoted avoidance and suppressed innate immunity during reinfection with bacteria, which we revealed to be specific to the previously exposed pathogens. During this trade-off switch of avoidance and innate immunity, the chemosensory neurons AWB and ADF modulated production of serotonin and dopamine, which in turn decreased expression of the innate immunity-associated genes and led to enhanced avoidance via the downstream insulin-like pathway. Therefore, our current study profiles the immune memories during C. elegans reinfected by pathogenic bacteria and further reveals that the chemosensory neurons, the neurotransmitter(s), and their associated molecular signaling pathways are responsible for a trade-off switch between the two immunological memories.
Collapse
Affiliation(s)
- Jinyuan Yan
- State Key Lab for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China; Center Laboratory of the Second Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Ninghui Zhao
- Neurosurgery of the Second Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Zhongshan Yang
- Faculty of Basic Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Yuhong Li
- State Key Lab for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Hua Bai
- State Key Lab for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China; School of Public Health, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Zou
- School of Public Health, Kunming Medical University, Kunming, Yunnan, China
| | - Keqin Zhang
- State Key Lab for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Xiaowei Huang
- State Key Lab for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China.
| |
Collapse
|
43
|
Nunes C, Sucena É, Koyama T. Endocrine regulation of immunity in insects. FEBS J 2020; 288:3928-3947. [PMID: 33021015 DOI: 10.1111/febs.15581] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Organisms have constant contact with potentially harmful agents that can compromise their fitness. However, most of the times these agents fail to cause serious disease by virtue of the rapid and efficient immune responses elicited in the host that can range from behavioural adaptations to immune system triggering. The immune system of insects does not comprise the adaptive arm, making it less complex than that of vertebrates, but key aspects of the activation and regulation of innate immunity are conserved across different phyla. This is the case for the hormonal regulation of immunity as a part of the broad organismal responses to external conditions under different internal states. In insects, depending on the physiological circumstances, distinct hormones either enhance or suppress the immune response integrating individual (and often collective) responses physiologically and behaviourally. In this review, we provide an overview of our current knowledge on the endocrine regulation of immunity in insects, its mechanisms and implications on metabolic adaptation and behaviour. We highlight the importance of this multilayered regulation of immunity in survival and reproduction (fitness) and its dependence on the hormonal integration with other mechanisms and life-history traits.
Collapse
Affiliation(s)
| | - Élio Sucena
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Coates LC, Mahoney J, Ramsey JS, Warwick E, Johnson R, MacCoss MJ, Krasnoff SB, Howe KJ, Moulton K, Saha S, Mueller LA, Hall DG, Shatters RG, Heck ML, Slupsky CM. Development on Citrus medica infected with 'Candidatus Liberibacter asiaticus' has sex-specific and -nonspecific impacts on adult Diaphorina citri and its endosymbionts. PLoS One 2020; 15:e0239771. [PMID: 33022020 PMCID: PMC7537882 DOI: 10.1371/journal.pone.0239771] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Huanglongbing (HLB) is a deadly, incurable citrus disease putatively caused by the unculturable bacterium, 'Candidatus Liberibacter asiaticus' (CLas), and transmitted by Diaphorina citri. Prior studies suggest D. citri transmits CLas in a circulative and propagative manner; however, the precise interactions necessary for CLas transmission remain unknown, and the impact of insect sex on D. citri-CLas interactions is poorly understood despite reports of sex-dependent susceptibilities to CLas. We analyzed the transcriptome, proteome, metabolome, and microbiome of male and female adult D. citri reared on healthy or CLas-infected Citrus medica to determine shared and sex-specific responses of D. citri and its endosymbionts to CLas exposure. More sex-specific than shared D. citri responses to CLas were observed, despite there being no difference between males and females in CLas density or relative abundance. CLas exposure altered the abundance of proteins involved in immunity and cellular and oxidative stress in a sex-dependent manner. CLas exposure impacted cuticular proteins and enzymes involved in chitin degradation, as well as energy metabolism and abundance of the endosymbiont 'Candidatus Profftella armatura' in both sexes similarly. Notably, diaphorin, a toxic Profftella-derived metabolite, was more abundant in both sexes with CLas exposure. The responses reported here resulted from a combination of CLas colonization of D. citri as well as the effect of CLas infection on C. medica. Elucidating these impacts on D. citri and their endosymbionts contributes to our understanding of the HLB pathosystem and identifies the responses potentially critical to limiting or promoting CLas acquisition and propagation in both sexes.
Collapse
Affiliation(s)
- Laurynne C. Coates
- Department of Food Science and Technology, University of California, Davis, California, United States of America
| | - Jaclyn Mahoney
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - John S. Ramsey
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
- Robert W. Holley Center for Agriculture and Health, Emerging Pests and Pathogens Research Unit, USDA Agricultural Research Service, Ithaca, New York, United States of America
| | - EricaRose Warwick
- Plant Pathology, University of Florida Citrus Research and Education Center, Lake Alfred, Florida, United States of America
| | - Richard Johnson
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Michael J. MacCoss
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Stuart B. Krasnoff
- Robert W. Holley Center for Agriculture and Health, Emerging Pests and Pathogens Research Unit, USDA Agricultural Research Service, Ithaca, New York, United States of America
| | - Kevin J. Howe
- Robert W. Holley Center for Agriculture and Health, Emerging Pests and Pathogens Research Unit, USDA Agricultural Research Service, Ithaca, New York, United States of America
| | - Kathy Moulton
- U.S. Horticultural Research Laboratory, Unit of Subtropical Insects and Horticulture, USDA Agricultural Research Service, Fort Pierce, Florida, United States of America
| | - Surya Saha
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - Lukas A. Mueller
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
| | - David G. Hall
- U.S. Horticultural Research Laboratory, Unit of Subtropical Insects and Horticulture, USDA Agricultural Research Service, Fort Pierce, Florida, United States of America
| | - Robert G. Shatters
- U.S. Horticultural Research Laboratory, Unit of Subtropical Insects and Horticulture, USDA Agricultural Research Service, Fort Pierce, Florida, United States of America
| | - Michelle L. Heck
- Boyce Thompson Institute for Plant Research, Ithaca, New York, United States of America
- Robert W. Holley Center for Agriculture and Health, Emerging Pests and Pathogens Research Unit, USDA Agricultural Research Service, Ithaca, New York, United States of America
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, New York, United States of America
| | - Carolyn M. Slupsky
- Department of Food Science and Technology, University of California, Davis, California, United States of America
| |
Collapse
|
45
|
González-González A, Wayne ML. Immunopathology and immune homeostasis during viral infection in insects. Adv Virus Res 2020; 107:285-314. [PMID: 32711732 DOI: 10.1016/bs.aivir.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Organisms clear infections by mounting an immune response that is normally turned off once the pathogens have been cleared. However, sometimes this immune response is not properly or timely arrested, resulting in the host damaging itself. This immune dysregulation may be referred to as immunopathology. While our knowledge of immune and metabolic pathways in insects, particularly in response to viral infections, is growing, little is known about the mechanisms that regulate this immune response and hence little is known about immunopathology in this important and diverse group of organisms. In this chapter we focus both on documenting the molecular mechanisms described involved in restoring immune homeostasis in insects after viral infections and on identifying potential mechanisms for future investigation. We argue that learning about the immunopathological consequences of an improperly regulated immune response in insects will benefit both insect and human health.
Collapse
Affiliation(s)
| | - Marta L Wayne
- Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
46
|
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
|
47
|
Tafesh-Edwards G, Eleftherianos I. JNK signaling in Drosophila immunity and homeostasis. Immunol Lett 2020; 226:7-11. [PMID: 32598968 DOI: 10.1016/j.imlet.2020.06.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/12/2020] [Accepted: 06/24/2020] [Indexed: 01/29/2023]
Abstract
As members of the mitogen-activated protein kinase (MAPK) family, the c-Jun N-terminal kinases (JNKs) regulate cell responses to a wide range of extrinsic and intrinsic insults, including irradiation, reactive oxygen species (ROS), DNA damage, heat, bacterial antigens, and inflammatory cytokines. Particularly, JNK signaling regulates and promotes many important physiological processes that influence metabolic and tissue homeostasis, cell death/survival, and cell damage repair and ultimately impacts the lifespan of an organism. This diverse functionality causes a variety of tissue-specific and context-specific cellular responses, mediated by various cross talks between JNK and other cellular signaling pathways. Thus, highlighting its significance as a determinant of stress responses, JNK loss-of-function mutations have been implicated in a multitude of pathologies, including neurodegenerative diseases, diabetes, and cancer. Because JNK functions are specified in a context-dependent manner and can greatly vary, the underlying causes for these different outcomes remain largely unresolved despite the gained knowledge of many regulatory roles of JNK signaling during the past two decades. In Drosophila melanogaster, JNK signaling is conserved and required for immune responses, as well as the development for morphogenetic processes (embryonic dorsal closure and thorax closure). Therefore, Drosophila innate immunity provides the ideal model to understand the complex mechanisms underlying JNK activation and regulation. In the following, we review studies in Drosophila that highlight several mechanisms by which JNK signaling influences immunity and homeostasis.
Collapse
Affiliation(s)
- Ghada Tafesh-Edwards
- Infection and Innate Immunity Lab, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Science and Engineering Hall, 800 22nd Street NW, Washington DC, 20052, USA
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Lab, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Science and Engineering Hall, 800 22nd Street NW, Washington DC, 20052, USA.
| |
Collapse
|
48
|
Ma L, Liu L, Zhao Y, Yang L, Chen C, Li Z, Lu Z. JNK pathway plays a key role in the immune system of the pea aphid and is regulated by microRNA-184. PLoS Pathog 2020; 16:e1008627. [PMID: 32584915 PMCID: PMC7343183 DOI: 10.1371/journal.ppat.1008627] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/08/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Different from holometabolous insects, the hemipteran species such as pea aphid Acyrthosiphon pisum exhibit reduced immune responses with the absence of the genes coding for antimicrobial peptide (AMP), immune deficiency (IMD), peptidoglycan recognition proteins (PGRPs), and other immune-related molecules. Prior studies have proved that phenoloxidase (PO)-mediated melanization, hemocyte-mediated phagocytosis, and reactive oxygen species (ROS) participate in pea aphid defense against bacterial infection. Also, the conserved signaling, Jun N-terminal kinase (JNK) pathway, has been suggested to be involved in pea aphid immune defense. However, the precise role of the JNK signaling, its interplay with other immune responses and its regulation in pea aphid are largely unknown. In this study, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway regulated hemolymph PO activity, hydrogen peroxide concentration and hemocyte phagocytosis in bacteria infected pea aphids, suggesting that the JNK pathway plays a central role in regulating immune responses in pea aphid. We further revealed the JNK pathway is regulated by microRNA-184 in response to bacterial infection. It is possible that in common the JNK pathway plays a key role in immune system of hemipteran insects and microRNA-184 regulates the JNK pathway in animals.
Collapse
Affiliation(s)
- Li Ma
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lu Liu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yujie Zhao
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Yang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Caihua Chen
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhaofei Li
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail:
| |
Collapse
|
49
|
Geigges M, Arekatla G, Paro R. Priming exposures to lipopolysaccharides do not affect the induction of Polycomb target genes upon re-exposure. PLoS One 2020; 15:e0231498. [PMID: 32287290 PMCID: PMC7156044 DOI: 10.1371/journal.pone.0231498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/24/2020] [Indexed: 11/18/2022] Open
Abstract
The Polycomb group (PcG) proteins are chromatin factors underlying the process of transcriptional memory to preserve developmental decisions and keep cellular identities. However, not only developmental signals need to be memorized and thus maintained during the life of an organism. For host protection against pathogens, also a memory of previous exposures to an immunogenic stimulus is crucial to mount a more protective immune response upon re-exposure. The antigen-specific adaptive immunity in vertebrates is an example of such a memory to previous immunogenic stimulation. Recently, adaptive characteristics were also attributed to innate immunity, which was classically seen to lack memory. However, the mechanistic details of an adaptive innate immune response are yet to be fully understood and chromatin-based epigenetic mechanisms seem to play an important role in this phenomenon. Possibly, PcG proteins can contribute to such an epigenetic innate immune memory. In this study, we analyzed whether the PcG system can mediate a transcriptional memory of exposure to lipopolysaccharides (LPS). To this end, various forms of LPS pre-treatment were applied to reporter cells and expression kinetics of PcG target genes were analyzed after a second LPS exposure. Neither single nor multiple LPS pre-treatment affected the induction of endogenous LPS-responsive transcripts upon re-exposure. Altogether, our extensive analyses did not provide any evidence for a PcG system-mediated memory of LPS stimulation.
Collapse
Affiliation(s)
- Marco Geigges
- Department of Biosystems Science and Engineering, Epigenomics Group, ETH Zurich, Basel, Switzerland
| | - Geethika Arekatla
- Department of Biosystems Science and Engineering, Epigenomics Group, ETH Zurich, Basel, Switzerland
| | - Renato Paro
- Department of Biosystems Science and Engineering, Epigenomics Group, ETH Zurich, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Sparks ME, Bansal R, Benoit JB, Blackburn MB, Chao H, Chen M, Cheng S, Childers C, Dinh H, Doddapaneni HV, Dugan S, Elpidina EN, Farrow DW, Friedrich M, Gibbs RA, Hall B, Han Y, Hardy RW, Holmes CJ, Hughes DST, Ioannidis P, Cheatle Jarvela AM, Johnston JS, Jones JW, Kronmiller BA, Kung F, Lee SL, Martynov AG, Masterson P, Maumus F, Munoz-Torres M, Murali SC, Murphy TD, Muzny DM, Nelson DR, Oppert B, Panfilio KA, Paula DP, Pick L, Poelchau MF, Qu J, Reding K, Rhoades JH, Rhodes A, Richards S, Richter R, Robertson HM, Rosendale AJ, Tu ZJ, Velamuri AS, Waterhouse RM, Weirauch MT, Wells JT, Werren JH, Worley KC, Zdobnov EM, Gundersen-Rindal DE. Brown marmorated stink bug, Halyomorpha halys (Stål), genome: putative underpinnings of polyphagy, insecticide resistance potential and biology of a top worldwide pest. BMC Genomics 2020; 21:227. [PMID: 32171258 PMCID: PMC7071726 DOI: 10.1186/s12864-020-6510-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/20/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Halyomorpha halys (Stål), the brown marmorated stink bug, is a highly invasive insect species due in part to its exceptionally high levels of polyphagy. This species is also a nuisance due to overwintering in human-made structures. It has caused significant agricultural losses in recent years along the Atlantic seaboard of North America and in continental Europe. Genomic resources will assist with determining the molecular basis for this species' feeding and habitat traits, defining potential targets for pest management strategies. RESULTS Analysis of the 1.15-Gb draft genome assembly has identified a wide variety of genetic elements underpinning the biological characteristics of this formidable pest species, encompassing the roles of sensory functions, digestion, immunity, detoxification and development, all of which likely support H. halys' capacity for invasiveness. Many of the genes identified herein have potential for biomolecular pesticide applications. CONCLUSIONS Availability of the H. halys genome sequence will be useful for the development of environmentally friendly biomolecular pesticides to be applied in concert with more traditional, synthetic chemical-based controls.
Collapse
Affiliation(s)
- Michael E Sparks
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA.
| | - Raman Bansal
- USDA-ARS San Joaquin Valley Agricultural Sciences Center, Parlier, CA, 93648, USA
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Michael B Blackburn
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Hsu Chao
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mengyao Chen
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Sammy Cheng
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | | | - Huyen Dinh
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Harsha Vardhan Doddapaneni
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shannon Dugan
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elena N Elpidina
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119911, Russia
| | - David W Farrow
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Richard A Gibbs
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Brantley Hall
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Yi Han
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard W Hardy
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Christopher J Holmes
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Daniel S T Hughes
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Panagiotis Ioannidis
- Department of Genetic Medicine and Development, University of Geneva Medical School and Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland
- Present address: Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 73100, Heraklion, Crete, Greece
| | | | - J Spencer Johnston
- Department of Entomology, Texas A&M University, College Station, TX, 77843, USA
| | - Jeffery W Jones
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Brent A Kronmiller
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA
| | - Faith Kung
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Sandra L Lee
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alexander G Martynov
- Center for Data-Intensive Biomedicine and Biotechnology, Skolkovo Institute of Science and Technology, Skolkovo, 143025, Russia
| | - Patrick Masterson
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Florian Maumus
- URGI, INRA, Université Paris-Saclay, 78026, Versailles, France
| | - Monica Munoz-Torres
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Shwetha C Murali
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Terence D Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Donna M Muzny
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David R Nelson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Brenda Oppert
- USDA-ARS Center for Grain and Animal Health Research, Manhattan, KS, 66502, USA
| | - Kristen A Panfilio
- Developmental Biology, Institute for Zoology: University of Cologne, 50674, Cologne, Germany
- School of Life Sciences, University of Warwick, Gibbet Hill Campus, Coventry, CV4 7AL, United Kingdom
| | - Débora Pires Paula
- EMBRAPA Genetic Resources and Biotechnology, Brasília, DF, 70770-901, Brazil
| | - Leslie Pick
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | | | - Jiaxin Qu
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Katie Reding
- Department of Entomology, University of Maryland, College Park, MD, 20742, USA
| | - Joshua H Rhoades
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Adelaide Rhodes
- Larner College of Medicine, The University of Vermont, Burlington, VT, 05452, USA
| | - Stephen Richards
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Present address: Earth BioGenome Project, University of California, Davis, Davis, CA, 95616, USA
| | - Rose Richter
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Hugh M Robertson
- Department of Entomology, University of Illinois, Urbana-Champaign, IL, 61801, USA
| | - Andrew J Rosendale
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Zhijian Jake Tu
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Arun S Velamuri
- USDA-ARS Invasive Insect Biocontrol and Behavior Laboratory, Beltsville, MD, 20705, USA
| | - Robert M Waterhouse
- Department of Ecology and Evolution, University of Lausanne and Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Matthew T Weirauch
- Division of Biomedical Informatics, and Division of Developmental Biology, Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Jackson T Wells
- Center for Genome Research and Biocomputing, Oregon State University, Corvallis, OR, 97331, USA
| | - John H Werren
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Kim C Worley
- Department of Human and Molecular Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Evgeny M Zdobnov
- Department of Genetic Medicine and Development, University of Geneva Medical School and Swiss Institute of Bioinformatics, 1211, Geneva, Switzerland
| | | |
Collapse
|