1
|
Perlmutter JI, Atadurdyyeva A, Schedl ME, Unckless RL. Wolbachia enhances the survival of Drosophila infected with fungal pathogens. BMC Biol 2025; 23:42. [PMID: 39934832 PMCID: PMC11817339 DOI: 10.1186/s12915-025-02130-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Wolbachia bacteria of arthropods are at the forefront of basic and translational research on multipartite host-symbiont-pathogen interactions. These vertically transmitted microbes are the most widespread endosymbionts on the planet due to factors including host reproductive manipulation and fitness benefits. Importantly, some strains of Wolbachia can inhibit viral pathogenesis within and between arthropod hosts. Mosquitoes carrying the wMel Wolbachia strain of Drosophila melanogaster have a greatly reduced capacity to spread viruses like dengue and Zika to humans. While significant research efforts have focused on viruses, relatively little attention has been given to Wolbachia-fungal interactions despite the ubiquity of fungal entomopathogens in nature. RESULTS Here, we demonstrate that Wolbachia increase the longevity of their Drosophila melanogaster hosts when challenged with a spectrum of yeast and filamentous fungal pathogens. We find that this pattern can vary based on host genotype, sex, and fungal species. Further, Wolbachia correlates with higher fertility and reduced pathogen titers during initial fungal infection, indicating a significant fitness benefit. Finally, RNA sequencing results show altered expression of many immune and stress response genes in the context of Wolbachia and fungal infection, suggesting host immunity may be involved in the mechanism. CONCLUSIONS This study demonstrates Wolbachia's protective role in diverse fungal pathogen interactions and determines that the phenotype is broad, but with several variables that influence both the presence and strength of the phenotype. It also is a critical step forward to understanding how symbionts can protect their hosts from a variety of pathogens.
Collapse
Affiliation(s)
| | - Aylar Atadurdyyeva
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Margaret E Schedl
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Robert L Unckless
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
2
|
Orlovskis Z, Singh A, Kliot A, Huang W, Hogenhout SA. The phytoplasma SAP54 effector acts as a molecular matchmaker for leafhopper vectors by targeting plant MADS-box factor SVP. eLife 2025; 13:RP98992. [PMID: 39763298 PMCID: PMC11706604 DOI: 10.7554/elife.98992] [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] [Indexed: 01/11/2025] Open
Abstract
Obligate parasites often trigger significant changes in their hosts to facilitate transmission to new hosts. The molecular mechanisms behind these extended phenotypes - where genetic information of one organism is manifested as traits in another - remain largely unclear. This study explores the role of the virulence protein SAP54, produced by parasitic phytoplasmas, in attracting leafhopper vectors. SAP54 is responsible for the induction of leaf-like flowers in phytoplasma-infected plants. However, we previously demonstrated that the insects were attracted to leaves and the leaf-like flowers were not required. Here, we made the surprising discovery that leaf exposure to leafhopper males is required for the attraction phenotype, suggesting a leaf response that distinguishes leafhopper sex in the presence of SAP54. In contrast, this phytoplasma effector alongside leafhopper females discourages further female colonization. We demonstrate that SAP54 effectively suppresses biotic stress response pathways in leaves exposed to the males. Critically, the host plant MADS-box transcription factor short vegetative phase (SVP) emerges as a key element in the female leafhopper preference for plants exposed to males, with SAP54 promoting the degradation of SVP. This preference extends to female colonization of male-exposed svp null mutant plants over those not exposed to males. Our research underscores the dual role of the phytoplasma effector SAP54 in host development alteration and vector attraction - integral to the phytoplasma life cycle. Importantly, we clarify how SAP54, by targeting SVP, heightens leaf vulnerability to leafhopper males, thus facilitating female attraction and subsequent plant colonization by the insects. SAP54 essentially acts as a molecular 'matchmaker', helping male leafhoppers more easily locate mates by degrading SVP-containing complexes in leaves. This study not only provides insights into the long reach of single parasite genes in extended phenotypes, but also opens avenues for understanding how transcription factors that regulate plant developmental processes intersect with and influence plant-insect interactions.
Collapse
Affiliation(s)
| | - Archana Singh
- John Innes Centre, Norwich Research ParkNorwichUnited Kingdom
| | - Adi Kliot
- John Innes Centre, Norwich Research ParkNorwichUnited Kingdom
| | - Weijie Huang
- John Innes Centre, Norwich Research ParkNorwichUnited Kingdom
| | | |
Collapse
|
3
|
Darby AM, Keith SA, Kalukin AA, Lazzaro BP. Chronic bacterial infections exert metabolic costs in Drosophila melanogaster. J Exp Biol 2025; 228:jeb249424. [PMID: 39801480 PMCID: PMC11832186 DOI: 10.1242/jeb.249424] [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: 08/11/2024] [Accepted: 11/12/2024] [Indexed: 02/19/2025]
Abstract
Bacterial infections can substantially impact host metabolic health as a result of the direct and indirect demands of sustaining an immune response and of nutrient piracy by the pathogen itself. Drosophila melanogaster and other insects that survive a sublethal bacterial infection often carry substantial pathogen burdens for the remainder of life. In this study, we asked whether these chronic infections exact metabolic costs for the host, and how these costs scale with the severity of chronic infection. We infected D. melanogaster with four bacterial species (Providencia rettgeri, Serratia marcescens, Enterococcus faecalis and Lactococcus lactis) and assayed metabolic traits in chronically infected survivors. We found that D. melanogaster carrying chronic infections were uniformly more susceptible to starvation than uninfected controls, and that sensitivity to starvation escalated with higher chronic pathogen burden. We observed some evidence for greater depletion of triglyceride and glycogen stores in D. melanogaster carrying chronic bacterial loads, although this varied among bacterial species. Chronically infected flies exhibit sustained upregulation of the immune response, which we hypothesized might contribute to the metabolic costs. Consistent with this prediction, genetic activation of the major innate immune signaling pathways depleted metabolic stores and increased starvation sensitivity even in the absence of infection. These results demonstrate that even sublethal infections can have substantial health and fitness consequences for the hosts, arising in part from pathogen-induced immune activation, and that the consequences scale quantitatively with the severity of infection.
Collapse
Affiliation(s)
- Andrea M. Darby
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA
| | - Scott A. Keith
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA
| | - Ananda A. Kalukin
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
4
|
Wang X, Bao H, Huang YC, Barua A, Lai CM, Sun J, Zhou Y, Cong F, Gong S, Chang CH, Deng WM. Sex-dimorphic tumor growth is regulated by tumor microenvironmental and systemic signals. SCIENCE ADVANCES 2024; 10:eads4229. [PMID: 39642218 PMCID: PMC11623276 DOI: 10.1126/sciadv.ads4229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/01/2024] [Indexed: 12/08/2024]
Abstract
Tumor growth and progression involve coordinated regulation by internal, microenvironmental, and systemic signals and often display conspicuous sexual dimorphism. The mechanisms governing the integration and coordination of these signals, along with their sex-based differences, remain largely unknown. Using a Drosophila tumor model originating from nonreproductive tissue, we show that female-biased tumor growth involves multifaceted communications among tumor cells, hemocytes, and neuroendocrine insulin-producing cells (IPCs). Notch-active tumor cells recruit hemocytes carrying the tumor necrosis factor-α (TNF-α) homolog Eiger to the tumor microenvironment (TME), activating the c-Jun N-terminal kinase (JNK) pathway in tumor cells, instigating the sexually dimorphic up-regulation of cytokine Unpaired 2 (Upd2). Upd2, in turn, exerts a distal influence by modulating the release of a Drosophila insulin-like peptide (Dilp2) from IPCs. Dilp2 then activates the insulin signaling in the tumor, thereby fostering sexual-dimorphic tumor growth. Together, these findings reveal a relay mechanism involving the TME and systemic signals that collectively control the sexual dimorphism of tumor growth.
Collapse
Affiliation(s)
- Xianfeng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Hongcun Bao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Anindita Barua
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | - Jie Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Youfang Zhou
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Fei Cong
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | | | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
5
|
Basu A, Tekade K, Singh A, Das PN, Prasad NG. Experimental evolution for improved postinfection survival selects for increased disease resistance in Drosophila melanogaster. Evolution 2024; 78:1831-1843. [PMID: 39212194 DOI: 10.1093/evolut/qpae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/23/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Disease resistance (defined as the host capacity to limit systemic infection intensity) and disease tolerance (defined as the host capacity to limit infection-induced damage) are 2 complementary defense strategies that help the hosts maximize their survival and fitness when infected with pathogens and parasites. In addition to the underlying physiological mechanisms, the existing theory postulates that these 2 strategies differ in terms of the conditions under which each strategy evolves in the host populations, their evolutionary dynamics, and the ecological and epidemiological consequences of their evolution. Here, we explored if one or both of these strategies evolve when host populations are subjected to selection for increased postinfection survival. We experimentally evolved Drosophila melanogaster populations, selecting for the flies that survived an infection with the entomopathogen Enterococcus faecalis. We found that the host populations evolved increased disease resistance in response to selection for increased survival. This was despite the physiological costs associated with increased resistance, the expression of which varied with the phase of infection. We did not find evidence of any change in disease tolerance in the evolved host populations.
Collapse
Affiliation(s)
- Aabeer Basu
- Evolutionary Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Kimaya Tekade
- Evolutionary Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Aparajita Singh
- Evolutionary Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Paresh Nath Das
- Evolutionary Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Nagaraj Guru Prasad
- Evolutionary Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
6
|
Mullinax SR, Darby AM, Gupta A, Chan P, Smith BR, Unckless RL. A suite of selective pressures supports the maintenance of alleles of a Drosophila immune peptide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.18.553899. [PMID: 37662279 PMCID: PMC10473621 DOI: 10.1101/2023.08.18.553899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The innate immune system provides hosts with a crucial first line of defense against pathogens. While immune genes are often among the fastest evolving genes in the genome, in Drosophila , antimicrobial peptides (AMPs) are notable exceptions. Instead, AMPs may be under balancing selection, such that over evolutionary timescales multiple alleles are maintained in populations. In this study, we focus on the Drosophila antimicrobial peptide Diptericin A, which has a segregating amino acid polymorphism associated with differential survival after infection with the Gram-negative bacteria Providencia rettgeri . Diptericin A also helps control opportunistic gut infections by common Drosophila gut microbes, especially those of Lactobacillus plantarum . In addition to genotypic effects on gut immunity, we also see strong sex-specific effects that are most prominent in flies without functional diptericin A . To further characterize differences in microbiomes between different diptericin genotypes, we used 16S metagenomics to look at the microbiome composition. We used both lab reared and wild caught flies for our sequencing and looked at overall composition as well as the differential abundance of individual bacterial families. Overall, we find flies that are homozygous for one allele of diptericin A are better equipped to survive a systemic infection from P. rettgeri , but in general have a shorter lifespans after being fed common gut commensals. Our results suggest a possible mechanism for the maintenance of genetic variation of diptericin A through the complex interactions of sex, systemic immunity, and the maintenance of the gut microbiome.
Collapse
|
7
|
Prakash A, Fenner F, Shit B, Salminen TS, Monteith KM, Khan I, Vale PF. IMD-mediated innate immune priming increases Drosophila survival and reduces pathogen transmission. PLoS Pathog 2024; 20:e1012308. [PMID: 38857285 PMCID: PMC11192365 DOI: 10.1371/journal.ppat.1012308] [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: 04/24/2024] [Revised: 06/21/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
Invertebrates lack the immune machinery underlying vertebrate-like acquired immunity. However, in many insects past infection by the same pathogen can 'prime' the immune response, resulting in improved survival upon reinfection. Here, we investigated the mechanistic basis and epidemiological consequences of innate immune priming in the fruit fly Drosophila melanogaster when infected with the gram-negative bacterial pathogen Providencia rettgeri. We find that priming in response to P. rettgeri infection is a long-lasting and sexually dimorphic response. We further explore the epidemiological consequences of immune priming and find it has the potential to curtail pathogen transmission by reducing pathogen shedding and spread. The enhanced survival of individuals previously exposed to a non-lethal bacterial inoculum coincided with a transient decrease in bacterial loads, and we provide strong evidence that the effect of priming requires the IMD-responsive antimicrobial-peptide Diptericin-B in the fat body. Further, we show that while Diptericin B is the main effector of bacterial clearance, it is not sufficient for immune priming, which requires regulation of IMD by peptidoglycan recognition proteins. This work underscores the plasticity and complexity of invertebrate responses to infection, providing novel experimental evidence for the effects of innate immune priming on population-level epidemiological outcomes.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Florence Fenner
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Tiina S. Salminen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katy M. Monteith
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Pedro F. Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
8
|
Perlmutter JI, Chapman JR, Wilkinson MC, Nevarez-Saenz I, Unckless RL. A single amino acid polymorphism in natural Metchnikowin alleles of Drosophila results in systemic immunity and life history tradeoffs. PLoS Genet 2024; 20:e1011155. [PMID: 38466751 PMCID: PMC10957085 DOI: 10.1371/journal.pgen.1011155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 03/21/2024] [Accepted: 01/26/2024] [Indexed: 03/13/2024] Open
Abstract
Antimicrobial peptides (AMPs) are at the interface of interactions between hosts and microbes and are therefore expected to be rapidly evolving in a coevolutionary arms race with pathogens. In contrast, previous work demonstrated that insect AMPs tend to evolve more slowly than the genome average. Metchikowin (Mtk) is a Drosophila AMP that has a single amino acid residue that segregates as either proline (P) or arginine (R) in populations of four different species, some of which diverged more than 10 million years ago. These results suggest that there is a distinct functional importance to each allele. The most likely hypotheses are driven by two main questions: does each allele have a different efficacy against different specific pathogens (specificity hypothesis)? Or, is one allele a more potent antimicrobial, but with a host fitness cost (autoimmune hypothesis)? To assess their functional differences, we created D. melanogaster lines with the P allele, R allele, or Mtk null mutation using CRISPR/Cas9 genome editing and performed a series of life history and infection assays to assess them. In males, testing of systemic immune responses to a repertoire of bacteria and fungi demonstrated that the R allele performs as well or better than the P and null alleles with most infections. Females show some results that contrast with males, with Mtk alleles either not contributing to survival or with the P allele outperforming the R allele. In addition, measurements of life history traits demonstrate that the R allele is more costly in the absence of infection for both sexes. These results are consistent with both the specificity hypothesis (either allele can perform better against certain pathogens depending on context), and the autoimmune hypothesis (the R allele is generally the more potent antimicrobial in males, and carries a fitness cost). These results provide strong in vivo evidence that differential fitness with or without infection and sex-based functional differences in alleles may be adaptive mechanisms of maintaining immune gene polymorphisms in contrast with expectations of rapid evolution. Therefore, a complex interplay of forces including pathogen species and host sex may lead to balancing selection for immune genotypes. Strikingly, this selection may act on even a single amino acid polymorphism in an AMP.
Collapse
Affiliation(s)
- Jessamyn I. Perlmutter
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Joanne R. Chapman
- Institute of Environmental and Scientific Research (ESR), Christchurch, New Zealand
| | - Mason C. Wilkinson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Isaac Nevarez-Saenz
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| | - Robert L. Unckless
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
| |
Collapse
|
9
|
Rai P, Webb EM, Kang L, Weger-Lucarelli J. Insulin reduces the transmission potential of chikungunya virus and activates the toll pathway in Aedes aegypti mosquitoes. INSECT MOLECULAR BIOLOGY 2023; 32:648-657. [PMID: 37334906 DOI: 10.1111/imb.12863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
Chikungunya virus (CHIKV) is an alphavirus that has re-emerged globally over the last two decades and has the potential to become endemic in the United States due to the presence of competent mosquito vectors, Aedes aegypti and Aedes albopictus. CHIK disease is characterised by fever, rash, and joint pain, and causes chronic debilitating joint pain and swelling in >50% of infected individuals. Given the disease severity caused by CHIKV and the global presence of vectors to facilitate its spread, strategies to reduce viral transmission are desperately needed; however, the human biological factors driving CHIKV transmission are poorly understood. Towards that end, we have previously shown that mosquitoes fed on alphavirus-infected obese mice have reduced infection and transmission rates compared to those fed on infected lean mice despite similar viremia in lean and obese mice. One of the many host factors that increase in obese hosts is insulin, which was previously shown to impact the infection of mosquitoes by several flaviviruses. However, insulin's impact on alphavirus infection of live mosquitoes is unknown and whether insulin influences mosquito-borne virus transmission has not been tested. To test this, we exposed A. aegypti mosquitoes to bloodmeals with CHIKV in the presence or absence of physiologically relevant levels of insulin and found that insulin significantly lowered both infection and transmission rates. RNA sequencing analysis on mosquito midguts isolated at 1-day-post-infectious-bloodmeal (dpbm) showed enrichment in genes in the Toll immune pathway in the presence of insulin, which was validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We then sought to determine if the Toll pathway plays a role in CHIKV infection of Ae. aegypti mosquitoes; therefore, we knocked down Myd88, a critical immune adaptor molecule for the Toll pathway, in live mosquitoes, and found increased CHIKV infection compared to the mock knockdown control group. Overall, these data demonstrate that insulin reduces CHIKV transmission by Ae. aegypti and activates the Toll pathway in mosquitoes, suggesting that conditions resulting in higher serum insulin concentrations may reduce alphavirus transmission. Finally, these studies suggest that strategies to activate insulin or Toll signalling in mosquitoes may be an effective control strategy against medically relevant alphaviruses.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| | - Emily M Webb
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
- Department of Entomology, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
10
|
Wang JB, Lu HL, Sheng H, St Leger RJ. A Drosophila melanogaster model shows that fast growing Metarhizium species are the deadliest despite eliciting a strong immune response. Virulence 2023; 14:2275493. [PMID: 37941391 PMCID: PMC10732690 DOI: 10.1080/21505594.2023.2275493] [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: 07/10/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
We used Drosophila melanogaster to investigate how differences between Metarhizium species in growth rate and mechanisms of pathogenesis influence the outcome of infection. We found that the most rapid germinators and growers in vitro and on fly cuticle were the fastest killers, suggesting that pre-penetration competence is key to Metarhizium success. Virulent strains also induced the largest immune response, which did not depend on profuse growth within hosts as virulent toxin-producing strains only proliferated post-mortem while slow-killing strains that were specialized to other insects grew profusely pre-mortem. Metarhizium strains have apparently evolved resistance to widely distributed defenses such as the defensin Toll product drosomycin, but they were inhibited by Bomanins only found in Drosophila spp. Disrupting a gene (Dif), that mediates Toll immunity has little impact on the lethality of most Metarhizium strains (an exception being the early diverged M. frigidum and another insect pathogen Beauveria bassiana). However, disrupting the sensor of fungal proteases (Persephone) allowed rapid proliferation of strains within hosts (with the exception of M. album), and flies succumbed rapidly. Persephone also mediates gender differences in immune responses that determine whether male or female flies die sooner. We conclude that some strain differences in growth within hosts depend on immune-mediated interactions but intrinsic differences in pathogenic mechanisms are more important. Thus, Drosophila varies greatly in tolerance to different Metarhizium strains, in part because some of them produce toxins. Our results further develop D. melanogaster as a tractable model system for understanding insect-Metarhizium interactions.
Collapse
Affiliation(s)
- Jonathan B Wang
- Department of Entomology, University of Maryland, College Park, MD, USA
| | - Hsiao-Ling Lu
- Department of Entomology, University of Maryland, College Park, MD, USA
| | - Huiyu Sheng
- Department of Entomology, University of Maryland, College Park, MD, USA
| | | |
Collapse
|
11
|
Malladi S, Sukkar D, Bonnefoy A, Falla-Angel J, Laval-Gilly P. Imidacloprid and acetamiprid synergistically downregulate spaetzle and myD88 of the Toll pathway in haemocytes of the European honeybee (Apis mellifera). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 104:104323. [PMID: 37995888 DOI: 10.1016/j.etap.2023.104323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Pollinator health has been of critical concern over the last few decades. The prevalence of the honeybee Colony Collapse Disorder (CCD), changing climate, and the rise of vector-borne honeybee diseases by Varroa destructor, have played a major role in the rapid decline of global honeybee populations. Honeybees are environmentally and economically significant actors in biodiversity. The impact of agricultural practices, such as pesticide use, has exacerbated the negative effects on honeybees. We demonstrate the synergistic effect of cocktails of the neonicotinoids imidacloprid and acetamiprid on honeybee haemocytes. Two genes responsible for critical immune responses, spaetzle and myD88, are consistently dysregulated following exposure to either neonicotinoid alone or as a mixture with or without an immune challenge. The 2018 ban of neonicotinoids in Europe, followed by the 2020 reauthorisation of imidacloprid in France and the current consideration to reinstate acetamiprid underscores the need to evaluate their cumulative impact on honeybee health.
Collapse
Affiliation(s)
| | - Dani Sukkar
- Université de Lorraine, INRAE, LSE, F-54000 Nancy, France; Université de Lorraine, IUT Thionville-Yutz, Plateforme de Recherche, Transfert de Technologie et Innovation (PRTI), 57970 Yutz, France
| | - Antoine Bonnefoy
- Université de Lorraine, IUT Thionville-Yutz, Plateforme de Recherche, Transfert de Technologie et Innovation (PRTI), 57970 Yutz, France
| | | | | |
Collapse
|
12
|
Koekemoer LL, Hajkazemian M, Zawada JW, Mirzaie M, Dahan-Moss YL, Emami SN. Data-driven networking of global transcriptomics and male sexual development in the main malaria vector, Anopheles funestus. Sci Rep 2023; 13:16798. [PMID: 37798302 PMCID: PMC10556010 DOI: 10.1038/s41598-023-43914-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023] Open
Abstract
Deaths from malaria remain staggering despite global support that drives research into new territories. One major gap is our understanding of the sexual biological aspects of the male mosquito, which maintain the vector population solidity. Although Anopheles funestus s.s. is an extremely efficient African vector, little is known about the network between its sexual physiology and gene expression. The Culicidae male's sexual maturity involves a suite of physiological changes, such as genitalia rotation that is necessary for successful mating to occur. We show that mating success is guided by genes and physiological plasticity. Transcriptome analysis between newly emerged males (immature) versus males with rotating genitalia (maturing) provides insight into possible molecular mechanisms regulating male sexual behaviour. Putative transcripts that were associated with male sexual maturation were identified and validated. The discovery of the functions of these transcripts could lead to identifying potential targets for innovative vector control interventions, and mosquito population suppression.
Collapse
Affiliation(s)
- L L Koekemoer
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, South Africa.
| | - M Hajkazemian
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - J W Zawada
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - M Mirzaie
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Y L Dahan-Moss
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases, a Division of the National Health Laboratory Service, Johannesburg, South Africa
| | - S N Emami
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
- Molecular Attraction AB, Elektravägen 10, Hägersten, 126 30, Stockholm, Sweden.
- Natural Resources Institute, FES, University of Greenwich, London, UK.
| |
Collapse
|
13
|
Arch M, Vidal M, Fuentes E, Abat AS, Cardona PJ. The reproductive status determines tolerance and resistance to Mycobacterium marinum in Drosophila melanogaster. Evol Med Public Health 2023; 11:332-347. [PMID: 37868078 PMCID: PMC10590161 DOI: 10.1093/emph/eoad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/27/2023] [Indexed: 10/24/2023] Open
Abstract
Sex and reproductive status of the host have a major impact on the immune response against infection. Our aim was to understand their impact on host tolerance or resistance in the systemic Mycobacterium marinum infection of Drosophila melanogaster. We measured host survival and bacillary load at time of death, as well as expression by quantitative real-time polymerase chain reaction of immune genes (diptericin and drosomycin). We also assessed the impact of metabolic and hormonal regulation in the protection against infection by measuring expression of upd3, impl2 and ecR. Our data showed increased resistance in actively mating flies and in mated females, while reducing their tolerance to infection. Data suggests that Toll and immune deficiency (Imd) pathways determine tolerance and resistance, respectively, while higher basal levels of ecR favours the stimulation of the Imd pathway. A dual role has been found for upd3 expression, linked to increased/decreased mycobacterial load at the beginning and later in infection, respectively. Finally, impl2 expression has been related to increased resistance in non-actively mating males. These results allow further assessment on the differences between sexes and highlights the role of the reproductive status in D. melanogaster to face infections, demonstrating their importance to determine resistance and tolerance against M. marinum infection.
Collapse
Affiliation(s)
- Marta Arch
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
| | - Maria Vidal
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Esther Fuentes
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
| | - Anmaw Shite Abat
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Department of Veterinary Paraclinical Studies, University of Gondar, Gondar, Ethiopia
| | - Pere-Joan Cardona
- Tuberculosis Research Unit, Germans Trias i Pujol Research Institute (IGTP), Badalona, Catalonia, Spain
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB), Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Catalonia, Spain
- Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Catalonia, Spain
- Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Catalonia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
14
|
Imrie RM, Walsh SK, Roberts KE, Lello J, Longdon B. Investigating the outcomes of virus coinfection within and across host species. PLoS Pathog 2023; 19:e1011044. [PMID: 37216391 DOI: 10.1371/journal.ppat.1011044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Interactions between coinfecting pathogens have the potential to alter the course of infection and can act as a source of phenotypic variation in susceptibility between hosts. This phenotypic variation may influence the evolution of host-pathogen interactions within host species and interfere with patterns in the outcomes of infection across host species. Here, we examine experimental coinfections of two Cripaviruses-Cricket Paralysis Virus (CrPV), and Drosophila C Virus (DCV)-across a panel of 25 Drosophila melanogaster inbred lines and 47 Drosophilidae host species. We find that interactions between these viruses alter viral loads across D. melanogaster genotypes, with a ~3 fold increase in the viral load of DCV and a ~2.5 fold decrease in CrPV in coinfection compared to single infection, but we find little evidence of a host genetic basis for these effects. Across host species, we find no evidence of systematic changes in susceptibility during coinfection, with no interaction between DCV and CrPV detected in the majority of host species. These results suggest that phenotypic variation in coinfection interactions within host species can occur independently of natural host genetic variation in susceptibility, and that patterns of susceptibility across host species to single infections can be robust to the added complexity of coinfection.
Collapse
Affiliation(s)
- Ryan M Imrie
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Sarah K Walsh
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Katherine E Roberts
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| | - Joanne Lello
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ben Longdon
- Centre for Ecology & Conservation, Faculty of Environment, Science, and Economy, Biosciences, University of Exeter, Penryn Campus, Penryn, United Kingdom
| |
Collapse
|
15
|
Wukitch AM, Lawrence MM, Satriale FP, Patel A, Ginder GM, Van Beek EJ, Gilani O, Chambers MC. Impact of Chronic Infection on Resistance and Tolerance to Secondary Infection in Drosophila melanogaster. Infect Immun 2023; 91:e0036022. [PMID: 36794959 PMCID: PMC10016074 DOI: 10.1128/iai.00360-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Prior exposure to a pathogen can greatly influence the outcome of a secondary infection, and although invertebrates lack classically defined adaptive immunity, their immune response is still influenced by prior immune challenges. While the strength and specificity of such immune priming depends highly on the host organism and infecting microbe, chronic bacterial infection of the fruit fly Drosophila melanogaster with species isolated from wild-caught fruit flies provides broad nonspecific protection against a later secondary bacterial infection. To determine how chronic infection influences progression of secondary infection, we specifically tested how chronic infection with Serratia marcescens and Enterococcus faecalis impacted both resistance and tolerance to a secondary infection with an unrelated bacterium, Providencia rettgeri, by simultaneously tracking survival and bacterial load postinfection across a range of infectious doses. We found that these chronic infections increased both tolerance and resistance to P. rettgeri. Further investigation of S. marcescens chronic infection also revealed robust protection against the highly virulent Providencia sneebia, and that protection was dependent on the initial infectious dose for S. marcescens with protective doses corresponding with significantly increased diptericin expression. While the increased expression of this antimicrobial peptide gene likely explains the increased resistance, increased tolerance is likely due to other alterations in organismal physiology, such as increased negative regulation of immunity or tolerance of ER stress. These findings provide a foundation for future studies on how chronic infection influences tolerance to secondary infection.
Collapse
Affiliation(s)
- Abigail M. Wukitch
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, USA
| | | | | | - Alexa Patel
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, USA
| | - Grace M. Ginder
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, USA
| | - Emily J. Van Beek
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, USA
| | - Owais Gilani
- Department of Mathematics, Bucknell University, Lewisburg, Pennsylvania, USA
| | - Moria C. Chambers
- Department of Biology, Bucknell University, Lewisburg, Pennsylvania, USA
| |
Collapse
|
16
|
Cross ST, Brehm AL, Dunham TJ, Rodgers CP, Keene AH, Borlee GI, Stenglein MD. Galbut Virus Infection Minimally Influences Drosophila melanogaster Fitness Traits in a Strain and Sex-Dependent Manner. Viruses 2023; 15:539. [PMID: 36851753 PMCID: PMC9965562 DOI: 10.3390/v15020539] [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: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Galbut virus (family Partitiviridae) infects Drosophila melanogaster and can be transmitted vertically from infected mothers or infected fathers with near perfect efficiency. This form of super-Mendelian inheritance should drive infection to 100% prevalence, and indeed, galbut virus is ubiquitous in wild D. melanogaster populations. However, on average, only about 60% of individual flies are infected. One possible explanation for this is that a subset of flies are resistant to infection. Although galbut virus-infected flies appear healthy, infection may be sufficiently costly to drive selection for resistant hosts, thereby decreasing overall prevalence. To test this hypothesis, we quantified a variety of fitness-related traits in galbut virus-infected flies from two lines from the Drosophila Genetic Reference Panel (DGRP). Galbut virus-infected flies had no difference in average lifespan and total offspring production compared to their uninfected counterparts. Galbut virus-infected DGRP-517 flies pupated and eclosed faster than their uninfected counterparts. Some galbut virus-infected flies exhibited altered sensitivity to viral, bacterial, and fungal pathogens. The microbiome composition of flies was not measurably perturbed by galbut virus infection. Differences in phenotype attributable to galbut virus infection varied as a function of fly sex and DGRP strain, and differences attributable to infection status were dwarfed by larger differences attributable to strain and sex. Thus, galbut virus infection does produce measurable phenotypic changes, with changes being minor, offsetting, and possibly net-negative.
Collapse
Affiliation(s)
- Shaun T. Cross
- Department of Environmental, Agricultural, and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ali L. Brehm
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Tillie J. Dunham
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Case P. Rodgers
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Alexandra H. Keene
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Grace I. Borlee
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark D. Stenglein
- Center for Vector-Borne and Infectious Diseases, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
17
|
Rai KE, Yin H, Bengo ALC, Cheek M, Courville R, Bagheri E, Ramezan R, Behseta S, Shahrestani P. Immune defense in Drosophila melanogaster depends on diet, sex, and mating status. PLoS One 2023; 18:e0268415. [PMID: 37053140 PMCID: PMC10101424 DOI: 10.1371/journal.pone.0268415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Immune defense is a complex trait that affects and is affected by many other host factors, including sex, mating, and dietary environment. We used the agriculturally relevant fungal emtomopathogen, Beauveria bassiana, and the model host organism Drosophila melanogaster to examine how the impacts of sex, mating, and dietary environment on immunity are interrelated. We showed that the direction of sexual dimorphism in immune defense depends on mating status and mating frequency. We also showed that post-infection dimorphism in immune defense changes over time and is affected by dietary condition both before and after infection. Supplementing the diet with protein-rich yeast improved post-infection survival but more so when supplementation was done after infection instead of before. The multi-directional impacts among immune defense, sex, mating, and diet are clearly complex, and while our study shines light on some of these relationships, further study is warranted. Such studies have potential downstream applications in agriculture and medicine.
Collapse
Affiliation(s)
- Kshama Ekanath Rai
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| | - Han Yin
- Department of Mathematics, California State University, Fullerton, Fullerton, California, United States of America
| | - Arnie Lynn C Bengo
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| | - Madison Cheek
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| | - Robert Courville
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| | - Elnaz Bagheri
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| | - Reza Ramezan
- Department of Statistic and Actuarial Science, University of Waterloo, Ontario, Canada
| | - Sam Behseta
- Department of Mathematics, California State University, Fullerton, Fullerton, California, United States of America
| | - Parvin Shahrestani
- Department of Biological Sciences, California State University, Fullerton, Fullerton, California, United States of America
| |
Collapse
|
18
|
Shit B, Prakash A, Sarkar S, Vale PF, Khan I. Ageing leads to reduced specificity of antimicrobial peptide responses in Drosophila melanogaster. Proc Biol Sci 2022; 289:20221642. [PMID: 36382522 PMCID: PMC9667363 DOI: 10.1098/rspb.2022.1642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
Evolutionary theory predicts a late-life decline in the force of natural selection, possibly leading to late-life deregulations of the immune system. A potential outcome of such deregulations is the inability to produce specific immunity against target pathogens. We tested this possibility by infecting multiple Drosophila melanogaster lines (with bacterial pathogens) across age groups, where either individual or different combinations of Imd- and Toll-inducible antimicrobial peptides (AMPs) were deleted using CRISPR gene editing. We show a high degree of non-redundancy and pathogen-specificity of AMPs in young flies: in some cases, even a single AMP could confer complete resistance. However, ageing led to drastic reductions in such specificity to target pathogens, warranting the action of multiple AMPs across Imd and Toll pathways. Moreover, use of diverse AMPs either lacked survival benefits or even accompanied survival costs post-infection. These features were also sexually dimorphic: females required a larger repertoire of AMPs than males but extracted equivalent survival benefits. Finally, age-specific expansion of the AMP-repertoire was accompanied with ageing-induced downregulation of negative-regulators of the Imd pathway and damage to renal function post-infection, as features of poorly regulated immunity. Overall, we could highlight the potentially non-adaptive role of ageing in producing less-specific AMP responses, across sexes and pathogens.
Collapse
Affiliation(s)
- Biswajit Shit
- Ashoka University, Plot No. 2, Rajiv Gandhi Education City, National Capital Region P.O. Rai, Sonepat, Haryana-131029, India
| | - Arun Prakash
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Saubhik Sarkar
- Ashoka University, Plot No. 2, Rajiv Gandhi Education City, National Capital Region P.O. Rai, Sonepat, Haryana-131029, India
| | - Pedro F. Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Imroze Khan
- Ashoka University, Plot No. 2, Rajiv Gandhi Education City, National Capital Region P.O. Rai, Sonepat, Haryana-131029, India
| |
Collapse
|
19
|
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: 7] [Impact Index Per Article: 2.3] [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
|
20
|
Corbally MK, Regan JC. Fly immunity comes of age: The utility of Drosophila as a model for studying variation in immunosenescence. FRONTIERS IN AGING 2022; 3:1016962. [PMID: 36268532 PMCID: PMC9576847 DOI: 10.3389/fragi.2022.1016962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Affiliation(s)
| | - Jennifer C. Regan
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Infection increases activity via Toll dependent and independent mechanisms in Drosophila melanogaster. PLoS Pathog 2022; 18:e1010826. [PMID: 36129961 PMCID: PMC9529128 DOI: 10.1371/journal.ppat.1010826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 10/03/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Host behavioural changes are among the most apparent effects of infection. ‘Sickness behaviour’ can involve a variety of symptoms, including anorexia, depression, and changed activity levels. Here, using a real-time tracking and behavioural profiling platform, we show that in Drosophila melanogaster, several systemic bacterial infections cause significant increases in physical activity, and that the extent of this activity increase is a predictor of survival time in some lethal infections. Using multiple bacteria and D. melanogaster immune and activity mutants, we show that increased activity is driven by at least two different mechanisms. Increased activity after infection with Micrococcus luteus, a Gram-positive bacterium rapidly cleared by the immune response, strictly requires the Toll ligand spätzle. In contrast, increased activity after infection with Francisella novicida, a Gram-negative bacterium that cannot be cleared by the immune response, is entirely independent of both Toll and the parallel IMD pathway. The existence of multiple signalling mechanisms by which bacterial infections drive increases in physical activity implies that this effect may be an important aspect of the host response. Sickness behaviours are often observed during infection. Animals have been shown to change their feeding, mating, social and resting (sleeping) behaviours in response to infection. We show here that fruit-flies infected with bacteria respond by increasing their physical activity and decreasing the amount of time spent sleeping. This increase in activity is seen in some, but not all, bacterial infections, and appears to be driven by at least two different mechanisms: with some bacteria, activating the immune response is the only requirement to induce increased activity, while other bacteria induce increased activity independently of known immune detection pathways. The biological role of increased activity is unclear; flies in the wild may be driven to flee sites where infection risk or pathogen burden is high. Alternatively, increased activity could serve a less direct anti-microbial function. For example, active animals may be more likely to encounter potential mates or food resource.
Collapse
|
22
|
Lipopolysaccharide -mediated resistance to host antimicrobial peptides and hemocyte-derived reactive-oxygen species are the major Providencia alcalifaciens virulence factors in Drosophila melanogaster. PLoS Pathog 2022; 18:e1010825. [PMID: 36084158 PMCID: PMC9491580 DOI: 10.1371/journal.ppat.1010825] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/21/2022] [Accepted: 08/23/2022] [Indexed: 02/07/2023] Open
Abstract
Bacteria from the genus Providencia are ubiquitous Gram-negative opportunistic pathogens, causing “travelers’ diarrhea”, urinary tract, and other nosocomial infections in humans. Some Providencia strains have also been isolated as natural pathogens of Drosophila melanogaster. Despite clinical relevance and extensive use in Drosophila immunity research, little is known about Providencia virulence mechanisms and the corresponding insect host defenses. To close this knowledge gap, we investigated the virulence factors of a representative Providencia species—P. alcalifaciens which is highly virulent to fruit flies and amenable to genetic manipulations. We generated a P. alcalifaciens transposon mutant library and performed an unbiased forward genetics screen in vivo for attenuated mutants. Our screen uncovered 23 mutants with reduced virulence. The vast majority of them had disrupted genes linked to lipopolysaccharide (LPS) synthesis or modifications. These LPS mutants were sensitive to cationic antimicrobial peptides (AMPs) in vitro and their virulence was restored in Drosophila mutants lacking most AMPs. Thus, LPS-mediated resistance to host AMPs is one of the virulence strategies of P. alcalifaciens. Another subset of P. alcalifaciens attenuated mutants exhibited increased susceptibility to reactive oxygen species (ROS) in vitro and their virulence was rescued by chemical scavenging of ROS in flies prior to infection. Using genetic analysis, we found that the enzyme Duox specifically in hemocytes is the source of bactericidal ROS targeting P. alcalifaciens. Consistently, the virulence of ROS-sensitive P. alcalifaciens mutants was rescued in flies with Duox knockdown in hemocytes. Therefore, these genes function as virulence factors by helping bacteria to counteract the ROS immune response. Our reciprocal analysis of host-pathogen interactions between D. melanogaster and P. alcalifaciens identified that AMPs and hemocyte-derived ROS are the major defense mechanisms against P. alcalifaciens, while the ability of the pathogen to resist these host immune responses is its major virulence mechanism. Thus, our work revealed a host-pathogen conflict mediated by ROS and AMPs. Pathogens express special molecules or structures called virulence factors to successfully infect a host. By identifying these factors, we can learn how hosts fight and how pathogens cause infections. Here, we identified virulence factors of the human and fruit fly pathogen Providencia alcalifaciens, by infecting flies with a series of mutants of this pathogen. In this way, we detected 23 mutants that were less virulent. Some of these less virulent mutants were hypersensitive to fruit fly immune defense molecules called antimicrobial peptides (AMPs), while others were sensitive to reactive oxygen species (ROS) produced by the immune cells. Notably, AMPs-sensitive mutants remained virulent in a Drosophila mutant that lacks AMPs, while pathogens sensitive to oxidative stress retained their virulence in a fruit fly mutant devoid of oxidative species. These results suggest that the ability of P. alcalifaciens to resist two major host immune molecules, namely AMPs and ROS, is the major virulence mechanism. Overall, our systematic analysis of P. alcalifaciens virulence factors has identified the major defense mechanisms of the fruit fly against this pathogen and the bacterial mechanisms to combat these immune responses.
Collapse
|
23
|
Duneau D, Ferdy JB. Pathogen within-host dynamics and disease outcome: what can we learn from insect studies? CURRENT OPINION IN INSECT SCIENCE 2022; 52:100925. [PMID: 35489681 DOI: 10.1016/j.cois.2022.100925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 06/14/2023]
Abstract
Parasite proliferations within/on the host form the basis of the outcome of all infectious diseases. However, within-host dynamics are difficult to study in vertebrates, as it requires regularly following pathogen proliferation from the start of the infection and at the organismal level. Invertebrate models allow for this monitoring under controlled conditions using population approaches. These approaches offer the possibility to describe many parameters of the within-host dynamics, such as rate of proliferation, probability to control the infection, and average time at which the pathogen is controlled. New parameters such as the Pathogen Load Upon Death and the Set-Point Pathogen Load have emerged to characterize within-host dynamics and better understand disease outcome. While contextualizing the potential of studying within-host dynamics in insects to build fundamental knowledge, we review what we know about within-host dynamics using insect models, and what it can offer to our knowledge of infectious diseases.
Collapse
Affiliation(s)
- David Duneau
- Université Toulouse 3 Paul Sabatier, CNRS, IRD, UMR5174 EDB (Laboratoire Évolution & Diversité Biologique), Toulouse, France; Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780 Oeiras, Portugal.
| | - Jean-Baptiste Ferdy
- Université Toulouse 3 Paul Sabatier, CNRS, IRD, UMR5174 EDB (Laboratoire Évolution & Diversité Biologique), Toulouse, France.
| |
Collapse
|
24
|
Deng D, Xing S, Liu X, Ji Q, Zhai Z, Peng W. Transcriptome analysis of sex-biased gene expression in the spotted-wing Drosophila, Drosophila suzukii (Matsumura). G3 GENES|GENOMES|GENETICS 2022; 12:6588685. [PMID: 35587603 PMCID: PMC9339319 DOI: 10.1093/g3journal/jkac127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022]
Abstract
Sexual dimorphism occurs widely throughout insects and has profound influences on evolutionary path. Sex-biased genes are considered to account for most of phenotypic differences between sexes. In order to explore the sex-biased genes potentially associated with sexual dimorphism and sexual development in Drosophila suzukii, a major devastating and invasive crop pest, we conducted whole-organism transcriptome profiling and sex-biased gene expression analysis on adults of both sexes. We identified transcripts of genes involved in several sex-specific physiological and functional processes, including transcripts involved in sex determination, reproduction, olfaction, and innate immune signals. A total of 11,360 differentially expressed genes were identified in the comparison, and 1,957 differentially expressed genes were female-biased and 4,231 differentially expressed genes were male-biased. The pathway predominantly enriched for differentially expressed genes was related to spliceosome, which might reflect the differences in the alternative splicing mechanism between males and females. Twenty-two sex determination and 16 sex-related reproduction genes were identified, and expression pattern analysis revealed that the majority of genes were differentially expressed between sexes. Additionally, the differences in sex-specific olfactory and immune processes were analyzed and the sex-biased expression of these genes may play important roles in pheromone and odor detection, and immune response. As a valuable dataset, our sex-specific transcriptomic data can significantly contribute to the fundamental elucidation of the molecular mechanisms of sexual dimorphism in fruit flies, and may provide candidate genes potentially useful for the development of genetic sexing strains, an important tool for sterile insect technique applications against this economically important species.
Collapse
Affiliation(s)
- Dan Deng
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University , Changsha 410081, China
| | - Shisi Xing
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University , Changsha 410081, China
| | - Xuxiang Liu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Lab of Biopesticide and Chemical Biology, Ministry of Education, Institute of Biological Control, Fujian Agriculture and Forestry University , Fuzhou 350002, China
| | - Qinge Ji
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Lab of Biopesticide and Chemical Biology, Ministry of Education, Institute of Biological Control, Fujian Agriculture and Forestry University , Fuzhou 350002, China
| | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University , Changsha 410081, China
| | - Wei Peng
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Hunan Normal University , Changsha 410081, China
| |
Collapse
|
25
|
Meisel RP, Asgari D, Schlamp F, Unckless RL. Induction and inhibition of Drosophila X chromosome gene expression are both impeded by the dosage compensation complex. G3 (BETHESDA, MD.) 2022; 12:6632659. [PMID: 35792851 PMCID: PMC9434221 DOI: 10.1093/g3journal/jkac165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/16/2022] [Indexed: 12/24/2022]
Abstract
Sex chromosomes frequently differ from the autosomes in the frequencies of genes with sexually dimorphic or tissue-specific expression. Multiple hypotheses have been put forth to explain the unique gene content of the X chromosome, including selection against male-beneficial X-linked alleles, expression limits imposed by the haploid dosage of the X in males, and interference by the dosage compensation complex on expression in males. Here, we investigate these hypotheses by examining differential gene expression in Drosophila melanogaster following several treatments that have widespread transcriptomic effects: bacterial infection, viral infection, and abiotic stress. We found that genes that are induced (upregulated) by these biotic and abiotic treatments are frequently under-represented on the X chromosome, but so are those that are repressed (downregulated) following treatment. We further show that whether a gene is bound by the dosage compensation complex in males can largely explain the paucity of both up- and downregulated genes on the X chromosome. Specifically, genes that are bound by the dosage compensation complex, or close to a dosage compensation complex high-affinity site, are unlikely to be up- or downregulated after treatment. This relationship, however, could partially be explained by a correlation between differential expression and breadth of expression across tissues. Nonetheless, our results suggest that dosage compensation complex binding, or the associated chromatin modifications, inhibit both up- and downregulation of X chromosome gene expression within specific contexts, including tissue-specific expression. We propose multiple possible mechanisms of action for the effect, including a role of Males absent on the first, a component of the dosage compensation complex, as a dampener of gene expression variance in both males and females. This effect could explain why the Drosophila X chromosome is depauperate in genes with tissue-specific or induced expression, while the mammalian X has an excess of genes with tissue-specific expression.
Collapse
Affiliation(s)
- Richard P Meisel
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204-5001, USA
| | - Danial Asgari
- Department of Biology and Biochemistry, University of Houston, 3455 Cullen Blvd, Houston, TX 77204-5001, USA
| | - Florencia Schlamp
- Department of Medicine, NYU Grossman School of Medicine, 435 E 30th St, New York, NY 10016, USA
| | - Robert L Unckless
- Department of Molecular Biosciences, University of Kansas, 4055 Haworth Hall, 1200 Sunnyside Avenue, Lawrence, KS 66045, USA
| |
Collapse
|
26
|
Xu P, Rice A, Li T, Wang J, Yang X, Yuan H, Graham RI, Wilson K. Partiti-like viruses from African armyworm increase larval and pupal mortality of a novel host: the Egyptian cotton leafworm. PEST MANAGEMENT SCIENCE 2022; 78:1529-1537. [PMID: 34965003 DOI: 10.1002/ps.6771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND The general principle of using microbes from one species to manage a different pest species has a clear precedent in the large-scale release of mosquitoes carrying a Wolbachia bacterium derived from Drosophila flies. New technologies will facilitate the discovery of microbes that can be used in a similar way. Previously, we found three novel partiti-like viruses in the African armyworm (Spodoptera exempta). To investigate further the utility and consistency of host shift of insect viruses as a potential pest management tool, we tested the interaction between the partiti-like viruses and another novel host, the Egyptian cotton leafworm (Spodoptera littoralis). RESULT We found that all three partiti-like viruses appeared to be harmful to the novel host S. littoralis, by causing increased larval and pupal mortality. No effect was observed on host fecundity, and partiti-like virus infection did not impact host susceptibility when challenged with another pathogen, the baculovirus SpliNPV. Transcriptome analysis of partiti-like virus-infected and noninfected S. littoralis indicated that the viruses could impact host gene-expression profiles of S. littoralis, but they impact different pathways to the two other Spodoptera species through effects on pathways related to immunity (Jak-STAT/Toll and Imd) and reproduction (insulin signaling/insect hormones). CONCLUSION Taken together with the previous findings in the novel host S. frugiperda, these results indicate a parasitic relationship between the partiti-like viruses and novel insect hosts, suggesting a possible use and novel pest management strategy through the artificial host shift of novel viruses. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Pengjun Xu
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, China
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| | - Annabel Rice
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| | - Tong Li
- Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Jie Wang
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, China
| | - Xianming Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - He Yuan
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | - Kenneth Wilson
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| |
Collapse
|
27
|
Wang Y, Zhang Q, Yu HZ. Functional analysis of a peptidoglycan recognition protein involved in the immune response in the common cutworm, Spodoptera litura. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 109:e21858. [PMID: 35289433 DOI: 10.1002/arch.21858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/26/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
Peptidoglycan recognition proteins (GRPs) are family of pattern recognition receptors (PRRs), which can recognize the peptidoglycan and trigger the innate immune system against the microorganisms in insects. In this study, we identified a GRP-LB from Spodoptera litura genome database and named SlGRP-LB, which contained a complete open reading frame (ORF) of 639 bp, encoding a protein of 212 amino acids with a signal peptide and GRP domain. Phylogenetic tree analysis suggested that the SlGRP-LB has a close relationship with Helicoverpa armigera GRP-LB (HaGRP-LB). Tissue expression analysis revealed that SlGRP-LB had a high expression level in the fat body. The expression levels of SlGRP-LB were significantly upregulated in the hemolymph, fat body, and midgut from 3 to 12 h after injection of Escherichia coli and Staphylococcus aureus, while the expression levels were not downregulated at 24 h postinfection. Knockdown of SlGRP-LB expression by RNA interference reduced the expression of antibacterial peptide-related genes in the fat body and midgut, while their expression levels were upregulated in the hemolymph. In addition, the recombinant SlGRP-LB was expressed by using E. coli expression system, and it exhibited binding activity to E. coli. Taken together, the data suggest that S. litura GRP-LB might play a crucial role in regulating immune response in S. litura.
Collapse
Affiliation(s)
- Ying Wang
- College of Life Sciences, Gannan Normal University, Ganzhou, People's Republic of China
- National Navel Orange Engineering Research Center, Ganzhou, People's Republic of China
| | - Qin Zhang
- College of Life Sciences, Gannan Normal University, Ganzhou, People's Republic of China
| | - Hai-Zhong Yu
- College of Life Sciences, Gannan Normal University, Ganzhou, People's Republic of China
- National Navel Orange Engineering Research Center, Ganzhou, People's Republic of China
| |
Collapse
|
28
|
Barletta Ferreira AB, Bahia AC, Pitaluga AN, Barros E, Gama dos Santos D, Bottino-Rojas V, Kubota MS, Oliveira PLD, Pimenta PFP, Traub-Csekö YM, Sorgine MHF. Sexual Dimorphism in Immune Responses and Infection Resistance in Aedes aegypti and Other Hematophagous Insect Vectors. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.847109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sexual dimorphism in immune function is prevalent across different species, where males trade their ability to fight pathogens for a practical reproductive function while females favor an extended lifespan. In insects, these differences in immune function reflect an evolutionary life strategy, where females have a presumably more robust immune system than insect males. Here, we evaluate immune functioning in four male and female insect vectors, Aedes aegypti (Diptera, Culicidae), Anopheles aquasalis (Diptera, Culicidae), Lutzomyia longipalpis (Diptera, Psychodidae) and Rhodnius prolixus (Hemiptera, Reduviidae). We show evidence that challenges the concept of immune sexual dimorphism in three of these insect vectors. In the three Diptera species, A. aegypti, A. aquasalis and L. longipalpis that transmit arboviruses, Plasmodium spp. (Haemospororida, Plasmodiidae) and Leishmania spp. (Trypanosomatida, Trypanosomatidae), respectively, unchallenged adult males express higher levels of immune-related genes than adult females and immature developmental stages. The main components of the Toll, IMD, and Jak/STAT pathways and antimicrobial effectors are highly expressed in whole-body males. Additionally, males present lower midgut basal microbiota levels than females. In A. aegypti mosquitoes, the differences in immune gene expression and microbiota levels are established in adult mosquitoes but are not present at the recently emerged adults and pupal stage. Antibiotic treatment does not affect the consistently higher expression of immune genes in males, except defensin, which is reduced significantly after microbiota depletion and restored after re-introduction. Our data suggest that Diptera males have a basal state of activation of the immune system and that activation of a more robust response through systemic immune challenge acutely compromises their survival. The ones who survive clear the infection entirely. Females follow a different strategy where a moderate immune reaction render higher tolerance to infection and survival. In contrast, hematophagous adult males of the Hemiptera vector R. prolixus, which transmits Trypanosoma cruzi, present no differences in immune activation compared to females, suggesting that diet differences between males and females may influence immune sexual dimorphism. These findings expand our understanding of the biology of insect vectors of human pathogens, which can help to direct the development of new strategies to limit vector populations.
Collapse
|
29
|
Specific Strains of Honeybee Gut Lactobacillus Stimulate Host Immune System to Protect against Pathogenic Hafnia alvei. Microbiol Spectr 2022; 10:e0189621. [PMID: 34985299 PMCID: PMC8729767 DOI: 10.1128/spectrum.01896-21] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Honeybee gut microbiota plays an important role in host physiology and metabolism. Recent studies have shown that the influence of the resident microorganisms in the regulation of honeybee immune system is profound, which protects against the pathogen Serratia marcescens. However, only few of the core gut members in the regulation of immune functions have been studied. Here, we explored how different bee gut bacterial species aided in the clearance of the pathogenic Hafnia alvei, which causes bee septicemia with a high mortality rate. We found that both Gilliamella apicola W8136 and Lactobacillus apis W8172 protect honeybees from the opportunistic pathogen, while two other strains from Gilliamella and Lactobacillus did not affect the invasion of H. alvei. Transcriptomic analysis revealed that gut species induced different expression profiles in the gut. Specifically, two regulator genes from the Toll pathway, PGRP-S3 recognizing Gram-positive and Spätzle that bind to the Toll protein for the downstream signal transduction, were elevated by L. apis. Correspondingly, multiple genes encoding antibacterial proteins were also stimulated by L. apis. Interestingly, we found an increased expression of apidaecin, which also exhibited a high in vitro inhibitory effect on H. alvei. To elucidate the difference of strains in the host’s immune regulation, comparative genomic analyses indicate that the S-layer proteins unique to L. apis are potentially involved in honeybee Toll signaling and the activation of antibacterial protein production. IMPORTANCE Honeybees are essential pollinators supporting global agricultural economies and food supplies. Recent honeybee decline has been linked to several factors, while pathogen infection is considered one of the most significant contributing factors. Although a limited number of bacterial pathogens have been identified, Hafnia alvei is one of the pathogens causing septicemia in adult bees. In this study, we showed that two bee gut members, Gilliamella and Lactobacillus, can clear H. alvei from invasion. Mono-colonization of specific strains can stimulate the host Toll signaling pathway and the downstream expression of AMPs. Specifically, apidaecin upregulated by the gut symbionts is more effective against the pathogen. Moreover, our genomic analysis suggests that the surface-layer proteins specific to Lactobacillus strains are an important driver of Toll signaling, highlighting the variation of bee gut strains in regulating the host immune system.
Collapse
|
30
|
Hrdina A, Iatsenko I. The roles of metals in insect-microbe interactions and immunity. CURRENT OPINION IN INSECT SCIENCE 2022; 49:71-77. [PMID: 34952239 DOI: 10.1016/j.cois.2021.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 06/14/2023]
Abstract
Metal ions play essential roles in diverse physiological processes in insects, including immunity and interactions with microbes. Some, like iron, are essential nutrients and therefore are the subject of a tug-of-war between insects and microbes. Recent findings showed that the hypoferremic response mediated by Transferrin 1 is an essential defense mechanism against pathogens in insects. Transferrin 1 and the overall iron metabolism were also implicated in mediating interactions between insects and beneficial microbes. Other metals, like copper and zinc, can interfere with insect immune effectors, and either enhance (antimicrobial peptides) or reduce (reactive oxygen species) their activity. By covering recent advances in the field, this review emphasizes the importance of metals as essential mediators of insect-microbe interactions.
Collapse
Affiliation(s)
- Alexandra Hrdina
- Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany
| | - Igor Iatsenko
- Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
31
|
Prakash A, Khan I. Why do insects evolve immune priming? A search for crossroads. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104246. [PMID: 34453994 DOI: 10.1016/j.dci.2021.104246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
Until recently, it was assumed that insects lack immune memory since they do not have vertebrate-like specialized memory cells. Therefore, their most well studied evolutionary response against pathogens was increased basal immunity. However, growing evidence suggests that many insects also exhibit a form of immune memory (immune priming), where prior exposure to a low dose of infection confers protection against subsequent infection by the same pathogen that acts both within and across generations. Most strikingly, they can rapidly evolve as a highly parallel and mutually exclusive strategy from basal immunity, under different selective conditions and with divergent evolutionary trade-offs. However, the relative importance of priming as an optimal immune strategy also has contradictions, primarily because supporting mechanisms are still unclear. In this review, we adopt a comparative approach to highlight several emerging evolutionary, ecological and mechanistic features of priming vs basal immune responses that warrant immediate attention for future research.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, EH9 3FL, United Kingdom.
| | - Imroze Khan
- Department of Biology, Ashoka University, Plot No. 2, Rajiv Gandhi Education City, P.O. Rai, Sonepat, Haryana, 131029, India.
| |
Collapse
|
32
|
Chen S, Chen L, Qi Y, Xu J, Ge Q, Fan Y, Chen D, Zhang Y, Wang L, Hou T, Yang X, Xi Y, Si J, Kang L, Wang L. Bifidobacterium adolescentis regulates catalase activity and host metabolism and improves healthspan and lifespan in multiple species. NATURE AGING 2021; 1:991-1001. [PMID: 37118342 DOI: 10.1038/s43587-021-00129-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 09/27/2021] [Indexed: 04/30/2023]
Abstract
To identify candidate bacteria associated with aging, we performed fecal microbiota sequencing in young, middle-aged and older adults, and found lower Bifidobacterium adolescentis abundance in older individuals aged ≥60 years. Dietary supplementation of B. adolescentis improved osteoporosis and neurodegeneration in a mouse model of premature aging (Terc-/-) and increased healthspan and lifespan in Drosophila melanogaster and Caenorhabditis elegans. B. adolescentis supplementation increased the activity of the catalase (CAT) enzyme in skeletal muscle and brain tissue from Terc-/- mice, and suppressed cellular senescence in mouse embryonic fibroblasts. Transgenic deletion of catalase (ctl-2) in C. elegans abolished the effects of B. adolescentis on the lifespan and healthspan. B. adolescentis feeding also led to changes in oxidative stress-associated metabolites in Terc-/- mouse feces. These results suggest a role for B. adolescentis in improving the healthspan and lifespan through the regulation of CAT activity and host metabolism.
Collapse
Affiliation(s)
- Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Luyi Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Yadong Qi
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Jilei Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Qiwei Ge
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yuedan Fan
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China
| | - Du Chen
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China
| | - Yawen Zhang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Lan Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Tongyao Hou
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China
| | - Xiaohang Yang
- Institute of Genetics and Department of Genetics, Division of Human Reproduction and Developmental Genetics of the Women's Hospital, Zhejiang University, Zhejiang, China
| | - Yongmei Xi
- Institute of Genetics and Department of Genetics, Division of Human Reproduction and Developmental Genetics of the Women's Hospital, Zhejiang University, Zhejiang, China
| | - Jianmin Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang, China.
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China.
| | - Lijun Kang
- Department of Neurobiology and Department of Neurosurgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China.
| | - Liangjing Wang
- Prevention and Treatment Research Center for Senescent Disease, Zhejiang University School of Medicine, Zhejiang, China.
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China.
| |
Collapse
|
33
|
Abstract
Horizontal gene transfer (HGT) is a driving force of microbial evolution. The gut of animals acts as a potent reservoir for the lateral transfer of virulence, fitness, and antimicrobial resistance genes through plasmids. Reduced-complexity models for the examination of host-microbe interactions involved in plasmid transfer are greatly desired. Thus, this study identifies the use of Drosophila melanogaster as a model organism for the conjugation of plasmids of various incompatibility groups in the gut. Enterobacteriaceae conjugation pairs were identified in vitro and used for oral inoculation of the Drosophila gut. Flies were enumerated for the donor, recipient, and transconjugant populations. Each donor-recipient pair was observed to persist in fly guts for the duration of the experiment. Gut concentrations of the donors and recipients were significantly different between male and female flies, with females generally demonstrating increased concentrations. Furthermore, host genetics significantly altered the concentrations of donors and recipients. However, transconjugant concentrations were not affected by host sex or genetics and were detected only in the IncPε and IncI1 plasmid groups. This study demonstrates Drosophila melanogaster as a model for gut-mediated plasmid transfer. IMPORTANCE Microbial evolution in the gut of animals due to horizontal gene transfer (HGT) is of significant interest for microbial evolution as well as within the context of human and animal health. Microbial populations evolve within the host, and factors from the bacteria and host interact to regulate this evolution. However, little is currently known about how host and bacterial factors regulate plasmid-mediated HGT in the gut. This study demonstrates the use of Drosophila and the roles of sexual dimorphism as well as plasmid incompatibility groups in HGT in the gut.
Collapse
|
34
|
Imrie RM, Roberts KE, Longdon B. Between virus correlations in the outcome of infection across host species: Evidence of virus by host species interactions. Evol Lett 2021; 5:472-483. [PMID: 34621534 PMCID: PMC8484721 DOI: 10.1002/evl3.247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Virus host shifts are a major source of outbreaks and emerging infectious diseases, and predicting the outcome of novel host and virus interactions remains a key challenge for virus research. The evolutionary relationships between host species can explain variation in transmission rates, virulence, and virus community composition between hosts, but it is unclear if correlations exist between related viruses in infection traits across novel hosts. Here, we measure correlations in viral load of four Cripavirus isolates across experimental infections of 45 Drosophilidae host species. We find positive correlations between every pair of viruses tested, suggesting that some host clades show broad susceptibility and could act as reservoirs and donors for certain types of viruses. Additionally, we find evidence of virus by host species interactions, highlighting the importance of both host and virus traits in determining the outcome of virus host shifts. Of the four viruses tested here, those that were more closely related tended to be more strongly correlated, providing tentative evidence that virus evolutionary relatedness may be a useful proxy for determining the likelihood of novel virus emergence, which warrants further research.
Collapse
Affiliation(s)
- Ryan M. Imrie
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| | - Katherine E. Roberts
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| | - Ben Longdon
- Centre for Ecology and Conservation, Biosciences, College of Life and Environmental SciencesUniversity of ExeterPenrynTR10 9FEUnited Kingdom
| |
Collapse
|
35
|
Mank JE, Rideout EJ. Developmental mechanisms of sex differences: from cells to organisms. Development 2021; 148:272484. [PMID: 34647574 DOI: 10.1242/dev.199750] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Male-female differences in many developmental mechanisms lead to the formation of two morphologically and physiologically distinct sexes. Although this is expected for traits with prominent differences between the sexes, such as the gonads, sex-specific processes also contribute to traits without obvious male-female differences, such as the intestine. Here, we review sex differences in developmental mechanisms that operate at several levels of biological complexity - molecular, cellular, organ and organismal - and discuss how these differences influence organ formation, function and whole-body physiology. Together, the examples we highlight show that one simple way to gain a more accurate and comprehensive understanding of animal development is to include both sexes.
Collapse
Affiliation(s)
- Judith E Mank
- Department of Zoology, Biodiversity Research Centre, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada.,Biosciences, University of Exeter, Penryn Campus, Penryn TR10 9FE, UK
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
36
|
Disparate regulation of IMD signaling drives sex differences in infection pathology in Drosophila melanogaster. Proc Natl Acad Sci U S A 2021; 118:2026554118. [PMID: 34341118 PMCID: PMC8364183 DOI: 10.1073/pnas.2026554118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sex differences in infection outcome are a widely observed phenomenon. While it is known that biological sex can influence an animal’s response to infection, the mechanisms through which these differences emerge are less clear. Here, we describe a mechanism through which heightened regulation of the IMD signaling pathway by female—but not male—Drosophila melanogaster reduces the cost of immune activity at the expense of resistance to bacterial infection. Through the masculinization of the main organ responsible for antimicrobial peptide activity in the fly (fat body), this work demonstrates that this heightened immune regulation is mediated by sex-determining pathways. Male and female animals exhibit differences in infection outcomes. One possible source of sexually dimorphic immunity is the sex-specific costs of immune activity or pathology, but little is known about the independent effects of immune- versus microbe-induced pathology and whether these may differ for the sexes. Here, by measuring metabolic and physiological outputs in Drosophila melanogaster with wild-type and mutant immune responses, we test whether the sexes are differentially impacted by these various sources of pathology and identify a critical regulator of this difference. We find that the sexes exhibit differential immune activity but similar bacteria-derived metabolic pathology. We show that female-specific immune-inducible expression of PGRP-LB, a negative regulator of the immune deficiency (IMD) pathway, enables females to reduce immune activity in response to reductions in bacterial numbers. In the absence of PGRP-LB, females are more resistant to infection, confirming the functional importance of this regulation and suggesting that female-biased immune restriction comes at a cost.
Collapse
|
37
|
Liu H, Guo S, He Y, Shi Q, Yang M, You X. Toll protein family structure, evolution and response of the whiteleg shrimp (Litopenaeus vannamei) to exogenous iridescent virus. JOURNAL OF FISH DISEASES 2021; 44:1131-1145. [PMID: 33835515 DOI: 10.1111/jfd.13374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 01/26/2021] [Accepted: 03/26/2021] [Indexed: 06/12/2023]
Abstract
Whiteleg shrimp is a widely cultured crustacean, but frequent disease outbreaks have decreased production and caused significant losses. Toll-like receptors (TLRs) comprise a large innate immune family that is involved in the innate immune response. However, understanding of their regulatory mechanism is limited. In this study, PacBio sequencing and Illumina sequencing were applied to the gill and hepatopancreas tissues of whiteleg shrimp and an integrated transcript gene set was established. The upregulation of Toll1, Toll2 and Toll3 transcripts in the hepatopancreas tissue of whiteleg shrimp after iridescent virus infection implies that these proteins are involved in the immune response to the virus; simultaneously, the TRAF6 and relish transcripts in the Toll pathway were also upregulated, implying that the Toll pathway was activated. We predicted the three-dimensional structure of the five Toll proteins in whiteleg shrimp and humans and constructed a phylogenetic tree of the Toll protein family. In addition, there was a large discrepancy of Toll1 between invertebrates and vertebrates, presumably because of the loss of Toll1 protein sequence during the evolution process from invertebrates to vertebrates. Our research will improve the cognition of Toll protein family in invertebrates in terms of evolution, structure and function and provide theoretical guidance for researchers in this field.
Collapse
Affiliation(s)
- Hongtao Liu
- Hainan Provincial Key Laboratory of Tropical Maricultural Technologies, Hainan Academy of Ocean and Fisheries Sciences, Haikou, China
| | - Shengtao Guo
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Yugui He
- Hainan Provincial Key Laboratory of Tropical Maricultural Technologies, Hainan Academy of Ocean and Fisheries Sciences, Haikou, China
| | - Qiong Shi
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Mingqiu Yang
- Hainan Provincial Key Laboratory of Tropical Maricultural Technologies, Hainan Academy of Ocean and Fisheries Sciences, Haikou, China
| | - Xinxin You
- Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
38
|
Bagchi B, Corbel Q, Khan I, Payne E, Banerji D, Liljestrand-Rönn J, Martinossi-Allibert I, Baur J, Sayadi A, Immonen E, Arnqvist G, Söderhäll I, Berger D. Sexual conflict drives micro- and macroevolution of sexual dimorphism in immunity. BMC Biol 2021; 19:114. [PMID: 34078377 PMCID: PMC8170964 DOI: 10.1186/s12915-021-01049-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sexual dimorphism in immunity is believed to reflect sex differences in reproductive strategies and trade-offs between competing life history demands. Sexual selection can have major effects on mating rates and sex-specific costs of mating and may thereby influence sex differences in immunity as well as associated host-pathogen dynamics. Yet, experimental evidence linking the mating system to evolved sexual dimorphism in immunity are scarce and the direct effects of mating rate on immunity are not well established. Here, we use transcriptomic analyses, experimental evolution and phylogenetic comparative methods to study the association between the mating system and sexual dimorphism in immunity in seed beetles, where mating causes internal injuries in females. RESULTS We demonstrate that female phenoloxidase (PO) activity, involved in wound healing and defence against parasitic infections, is elevated relative to males. This difference is accompanied by concomitant sex differences in the expression of genes in the prophenoloxidase activating cascade. We document substantial phenotypic plasticity in female PO activity in response to mating and show that experimental evolution under enforced monogamy (resulting in low remating rates and reduced sexual conflict relative to natural polygamy) rapidly decreases female (but not male) PO activity. Moreover, monogamous females had evolved increased tolerance to bacterial infection unrelated to mating, implying that female responses to costly mating may trade off with other aspects of immune defence, an hypothesis which broadly accords with the documented sex differences in gene expression. Finally, female (but not male) PO activity shows correlated evolution with the perceived harmfulness of male genitalia across 12 species of seed beetles, suggesting that sexual conflict has a significant influence on sexual dimorphisms in immunity in this group of insects. CONCLUSIONS Our study provides insights into the links between sexual conflict and sexual dimorphism in immunity and suggests that selection pressures moulded by mating interactions can lead to a sex-specific mosaic of immune responses with important implications for host-pathogen dynamics in sexually reproducing organisms.
Collapse
Affiliation(s)
- Basabi Bagchi
- Department of Biology, Ashoka University, Sonipat, India
| | - Quentin Corbel
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
- Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
| | - Imroze Khan
- Department of Biology, Ashoka University, Sonipat, India
| | - Ellen Payne
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | | | - Johanna Liljestrand-Rönn
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Ivain Martinossi-Allibert
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Julian Baur
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Ahmed Sayadi
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
- Department of Chemistry, Biochemistry, Uppsala University, Uppsala, Sweden
| | - Elina Immonen
- Department of Ecology and Genetics, Program of Evolutionary Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Göran Arnqvist
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Irene Söderhäll
- Department of Organismal Biology, Program of Comparative Physiology, Uppsala University, Uppsala, Sweden
| | - David Berger
- Department of Ecology and Genetics, Program of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Millington JW, Rideout EJ. Sexual Dimorphism: Ecdysone Modulates Sex Differences in the Gut. Curr Biol 2021; 30:R1327-R1330. [PMID: 33142105 DOI: 10.1016/j.cub.2020.08.088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sex differences in the Drosophila gut were discovered only recently. Recent work significantly extends our understanding of how steroid hormones specify these male-female differences by revealing a key role for ecdysone in regulating intestinal stem cell proliferation.
Collapse
Affiliation(s)
- Jason W Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
40
|
Xu P, Yang L, Yang X, Li T, Graham RI, Wu K, Wilson K. Novel partiti-like viruses are conditional mutualistic symbionts in their normal lepidopteran host, African armyworm, but parasitic in a novel host, Fall armyworm. PLoS Pathog 2020; 16:e1008467. [PMID: 32569314 PMCID: PMC7332103 DOI: 10.1371/journal.ppat.1008467] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 07/02/2020] [Accepted: 03/08/2020] [Indexed: 11/18/2022] Open
Abstract
Recent advances in next generation sequencing (NGS) (e.g. metagenomic and transcriptomic sequencing) have facilitated the discovery of a large number of new insect viruses, but the characterization of these viruses is still in its infancy. Here, we report the discovery, using RNA-seq, of three new partiti-like viruses from African armyworm, Spodoptera exempta (Lepidoptera: Noctuidae), which are all vertically-transmitted transovarially from mother to offspring with high efficiency. Experimental studies show that the viruses reduce their host's growth rate and reproduction, but enhance their resistance to a nucleopolyhedrovirus (NPV). Via microinjection, these partiti-like viruses were transinfected into a novel host, a newly-invasive crop pest in sub-Saharan Africa (SSA), the Fall armyworm, S. frugiperda. This revealed that in this new host, these viruses appear to be deleterious without any detectable benefit; reducing their new host's reproductive rate and increasing their susceptibility to NPV. Thus, the partiti-like viruses appear to be conditional mutualistic symbionts in their normal host, S. exempta, but parasitic in the novel host, S. frugiperda. Transcriptome analysis of S. exempta and S. frugiperda infected, or not, with the partiti-like viruses indicates that the viruses may regulate pathways related to immunity and reproduction. These findings suggest a possible pest management strategy via the artificial host-shift of novel viruses discovered by NGS.
Collapse
Affiliation(s)
- Pengjun Xu
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, China
- Lancaster Environment Centre, Lancaster University, Lancaster, United Kingdom
| | - Liyu Yang
- Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, China
| | - Xianming Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tong Li
- Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Robert I. Graham
- Department of Animal and Agriculture, Hartpury University, Gloucester, United Kingdom
| | - Kongming Wu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kenneth Wilson
- Lancaster Environment Centre, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
41
|
Arcila F, Meunier J. Friend or foe? The apparent benefits of gregarine (Apicomplexa: Sporozoa) infection in the European earwig. Int J Parasitol 2020; 50:461-469. [PMID: 32224124 DOI: 10.1016/j.ijpara.2020.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 11/18/2022]
Abstract
Studying the costs and benefits of host-parasite interactions is of central importance to shed light on the evolutionary drivers of host life history traits. Although gregarines (Apicomplexa: Sporozoa) are one of the most frequent parasites in the gut of invertebrates, the diversity of its potential impacts on a host remains poorly explored. In this study, we addressed this gap in knowledge by investigating the prevalence of natural infections by the gregarine Gregarina ovata and testing how these infections shape a large set of morphological, behavioural and physiological traits in the European earwig Forficula auricularia. Our results first show that G. ovata was present in 76.8% of 573 field-sampled earwigs, and that its prevalence was both higher in males compared with females and increased between July and September. The load of G. ovata in the infected individuals was higher in males than females, but this sex difference vanished during the season. Our experiments then surprisingly revealed apparent benefits of G. ovata infections. Food-deprived hosts survived longer when they exhibited high compared with low gregarine loads. Moreover, the presence of gregarines was associated with a reduced phenoloxidase activity, indicating a lower immune resistance or a higher immune tolerance of the infected hosts. By contrast, we found no effect of G. ovata presence and number on earwigs' development (eye distance, forceps length), activity, food consumption or resistance against a fungal pathogen. Overall, our findings suggest that G. ovata could be involved in a mutualistic relationship with the European earwig. Given the ubiquitous presence of gregarines among invertebrates, our data also suggest that this common member of insect gut flora could have a broad and positive role in the life history of many host species.
Collapse
Affiliation(s)
- Francisco Arcila
- Institute of Organismic and Molecular Evolutionary Biology, Johannes-Gutenberg University of Mainz, Mainz, Germany
| | - Joël Meunier
- Institut de Recherche sur la Biologie de l'Insecte, UMR 7261, CNRS, Université de Tours, Tours, France.
| |
Collapse
|
42
|
The Genetic Basis of Natural Variation in Drosophila melanogaster Immune Defense against Enterococcus faecalis. Genes (Basel) 2020; 11:genes11020234. [PMID: 32098395 PMCID: PMC7074548 DOI: 10.3390/genes11020234] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/18/2020] [Accepted: 02/18/2020] [Indexed: 01/03/2023] Open
Abstract
Dissecting the genetic basis of natural variation in disease response in hosts provides insights into the coevolutionary dynamics of host-pathogen interactions. Here, a genome-wide association study of Drosophila melanogaster survival after infection with the Gram-positive entomopathogenic bacterium Enterococcus faecalis is reported. There was considerable variation in defense against E. faecalis infection among inbred lines of the Drosophila Genetics Reference Panel. We identified single nucleotide polymorphisms associated with six genes with a significant (p < 10-08, corresponding to a false discovery rate of 2.4%) association with survival, none of which were canonical immune genes. To validate the role of these genes in immune defense, their expression was knocked-down using RNAi and survival of infected hosts was followed, which confirmed a role for the genes krishah and S6k in immune defense. We further identified a putative role for the Bomanin gene BomBc1 (also known as IM23), in E. faecalis infection response. This study adds to the growing set of association studies for infection in Drosophila melanogaster and suggests that the genetic causes of variation in immune defense differ for different pathogens.
Collapse
|
43
|
Belmonte RL, Corbally MK, Duneau DF, Regan JC. Sexual Dimorphisms in Innate Immunity and Responses to Infection in Drosophila melanogaster. Front Immunol 2020; 10:3075. [PMID: 32076419 PMCID: PMC7006818 DOI: 10.3389/fimmu.2019.03075] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/16/2019] [Indexed: 12/20/2022] Open
Abstract
The sexes show profound differences in responses to infection and the development of autoimmunity. Dimorphisms in immune responses are ubiquitous across taxa, from arthropods to vertebrates. Drosophila melanogaster shows strong sex dimorphisms in immune system responses at baseline, upon pathogenic challenge, and over aging. We have performed an exhaustive survey of peer-reviewed literature on Drosophila immunity, and present a database of publications indicating the sex(es) analyzed in each study. While we found a growing interest in the community in adult immunity and in reporting both sexes, the main body of work in this field uses only one sex, or does not stratify by sex. We synthesize evidence for sexually dimorphic responses to bacterial, viral, and fungal infections. Dimorphisms may be mediated by distinct immune compartments, and we review work on sex differences in behavioral, epithelial, cellular, and systemic (fat body-mediated) immunity. Emerging work on sexually dimorphic aging of immune tissues, immune senescence, and inflammation are examined. We consider evolutionary drivers for sex differences in immune investment, highlight the features of Drosophila biology that make it particularly amenable to studies of immune dimorphisms, and discuss areas for future exploration.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Mary-Kate Corbally
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - David F. Duneau
- Laboratoire Evolution & Diversite Biologique, UMR5174 EDB, CNRS, Université Toulouse 3 Paul Sabatier, Toulouse, France
| | - Jennifer C. Regan
- Institute of Immunology & Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
44
|
Cardona PJ, Català M, Prats C. Origin of tuberculosis in the Paleolithic predicts unprecedented population growth and female resistance. Sci Rep 2020; 10:42. [PMID: 31913313 PMCID: PMC6949267 DOI: 10.1038/s41598-019-56769-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
Current data estimate the origin of Mycobacterium tuberculosis complex (MtbC) infection around 73,000 years before the common era (BCE), and its evolution to “modern” lineages around 46,000 BCE. Being MtbC a major killer of humanity, the question is how both species could persist. To answer this question, we have developed two new epidemiological models (SEIR type), adapted to sex dimorphism and comparing coinfection and superinfection for different MtbC lineages. We have attributed a higher resistance/tolerance to females to explain the lower incidence noted in this sex, a better health status in the Paleolithic compared to the Neolithic, and a higher dissemination of “modern” lineages compared to “ancient” ones. Our findings show the extraordinary impact caused by “modern” lineages, provoking the extinction of the groups infected. This could only be overcomed by an unprecedented population increase (x20 times in 100 years) and helped with the protection generated by previous infection with “ancient” lineages. Our findings also suggest a key role of female resistance against MtbC. This data obliges us to rethink the growth population parameters in the Paleolithic, which is crucial to understanding the survival of both MtbC and humans, and to decipher the nature of human female resistance against TB.
Collapse
Affiliation(s)
- Pere-Joan Cardona
- Unitat de Tuberculosi Experimental, Institut de Recerca Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, CIBERES, Badalona, Catalonia, Spain.
| | - Martí Català
- Comparative Medicine and Bioimage Centre of Catalonia (CMCiB). Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Clara Prats
- Escola Superior d'Agricultura de Barcelona, Departament de Física, Universitat Politècnica de Catalunya (UPC)-BarcelonaTech, Castelldefels, Catalonia, Spain
| |
Collapse
|
45
|
Chambers MC, Jacobson E, Khalil S, Lazzaro BP. Consequences of chronic bacterial infection in Drosophila melanogaster. PLoS One 2019; 14:e0224440. [PMID: 31648237 PMCID: PMC6812774 DOI: 10.1371/journal.pone.0224440] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/14/2019] [Indexed: 11/25/2022] Open
Abstract
Even when successfully surviving an infection, a host often fails to eliminate a pathogen completely and may sustain substantial pathogen burden for the remainder of its life. Using systemic bacterial infection in Drosophila melanogaster, we characterize chronic infection by three bacterial species from different genera - Providencia rettgeri, Serratia marcescens, and Enterococcus faecalis–following inoculation with a range of doses. To assess the consequences of these chronic infections, we determined the expression of antimicrobial peptide genes, survival of secondary infection, and starvation resistance after one week of infection. While higher infectious doses unsurprisingly lead to higher risk of death, they also result in higher chronic bacterial loads among the survivors for all three infections. All three chronic infections caused significantly elevated expression of antimicrobial peptide genes at one week post-infection and provided generalized protection again secondary bacterial infection. Only P. rettgeri infection significantly influenced resistance to starvation, with persistently infected flies dying more quickly under starvation conditions relative to controls. These results suggest that there is potentially a generalized mechanism of protection against secondary infection, but that other impacts on host physiology may depend on the specific pathogen. We propose that chronic infections in D. melanogaster could be a valuable tool for studying tolerance of infection, including impacts on host physiology and behavior.
Collapse
Affiliation(s)
- Moria Cairns Chambers
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Department of Biology, Bucknell University, Lewisburg, PA, United States of America
- * E-mail:
| | - Eliana Jacobson
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
| | - Sarah Khalil
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
| | - Brian P. Lazzaro
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
46
|
Lea JK, Unckless RL. An assessment of the immune costs associated with meiotic drive elements in Drosophila. Proc Biol Sci 2019; 286:20191534. [PMID: 31530140 PMCID: PMC6784720 DOI: 10.1098/rspb.2019.1534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022] Open
Abstract
Most organisms are constantly adapting to pathogens and parasites that exploit their host for their own benefit. Less studied, but perhaps more ubiquitous, are intragenomic parasites or selfish genetic elements. These include transposable elements, selfish B chromosomes and meiotic drivers that promote their own replication without regard to fitness effects on hosts. Therefore, intragenomic parasites are also a constant evolutionary pressure on hosts. Gamete-killing meiotic drive elements are often associated with large chromosomal inversions that reduce recombination between the drive and wild-type chromosomes. This reduced recombination is thought to reduce the efficacy of selection on the drive chromosome and allow for the accumulation of deleterious mutations. We tested whether gamete-killing meiotic drive chromosomes were associated with reduced immune defence against two bacterial pathogens in three species of Drosophila. We found little evidence of reduced immune defence in lines with meiotic drive. One line carrying the Drosophila melanogaster autosomal Segregation Distorter did show reduced defence, but we were unable to attribute that reduced defence to either genotype or immune gene expression differences. Our results suggest that though gamete-killing meiotic drive chromosomes probably accumulate deleterious mutations, those mutations do not result in reduced capacity for immune defence.
Collapse
Affiliation(s)
| | - Robert L. Unckless
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
47
|
Brown LD, Shapiro LLM, Thompson GA, Estévez‐Lao TY, Hillyer JF. Transstadial immune activation in a mosquito: Adults that emerge from infected larvae have stronger antibacterial activity in their hemocoel yet increased susceptibility to malaria infection. Ecol Evol 2019; 9:6082-6095. [PMID: 31161020 PMCID: PMC6540708 DOI: 10.1002/ece3.5192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 01/24/2023] Open
Abstract
Larval and adult mosquitoes mount immune responses against pathogens that invade their hemocoel. Although it has been suggested that a correlation exists between immune processes across insect life stages, the influence that an infection in the hemocoel of a larva has on the immune system of the eclosed adult remains unknown. Here, we used Anopheles gambiae to test whether a larval infection influences the adult response to a subsequent bacterial or malaria parasite infection. We found that for both female and male mosquitoes, a larval infection enhances the efficiency of bacterial clearance following a secondary infection in the hemocoel of adults. The adults that emerge from infected larvae have more hemocytes than adults that emerge from naive or injured larvae, and individual hemocytes have greater phagocytic activity. Furthermore, mRNA abundance of immune genes-such as cecropin A, Lysozyme C1, Stat-A, and Tep1-is higher in adults that emerge from infected larvae. A larval infection, however, does not have a meaningful effect on the probability that female adults will survive a systemic bacterial infection, and increases the susceptibility of females to Plasmodium yoelii, as measured by oocyst prevalence and intensity in the midgut. Finally, immune proficiency varies by sex; females exhibit increased bacterial killing, have twice as many hemocytes, and more highly express immune genes. Together, these results show that a larval hemocoelic infection induces transstadial immune activation-possibly via transstadial immune priming-but that it confers both costs and benefits to the emerged adults.
Collapse
Affiliation(s)
- Lisa D. Brown
- Department of Biological SciencesVanderbilt UniversityNashvilleTennessee
- Present address:
Department of BiologyGeorgia Southern UniversityStatesboroGeorgia
| | | | | | | | - Julián F. Hillyer
- Department of Biological SciencesVanderbilt UniversityNashvilleTennessee
| |
Collapse
|
48
|
Galenza A, Foley E. Immunometabolism: Insights from the Drosophila model. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 94:22-34. [PMID: 30684503 DOI: 10.1016/j.dci.2019.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
Multicellular organisms inhabit an environment that includes a mix of essential nutrients and large numbers of potentially harmful microbes. Germline-encoded receptors scan the environment for microbe associated molecular patterns, and, upon engagement, activate powerful defenses to protect the host from infection. At the same time, digestive enzymes and transporter molecules sieve through ingested material for building blocks and energy sources necessary for survival, growth, and reproduction. We tend to view immune responses as a potent array of destructive forces that overwhelm potentially harmful agents. In contrast, we view metabolic processes as essential, constructive elements in the maintenance and propagation of life. However, there is considerable evidence of functional overlap between the two processes, and disruptions to one frequently modify outputs of the other. Studies of immunometabolism, or interactions between immunity and metabolism, have increased in prominence with the discovery of inflammatory components to metabolic diseases such as type two diabetes. In this review, we will focus on contributions of studies with the fruit fly, Drosophila melanogaster, to our understanding of immunometabolism. Drosophila is widely used to study immune signaling, and to understand the regulation of metabolism in vivo, and this insect has considerable potential as a tool to build our understanding of the molecular and cellular bridges that connect immune and metabolic pathways.
Collapse
Affiliation(s)
- Anthony Galenza
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada.
| |
Collapse
|
49
|
Troha K, Buchon N. Methods for the study of innate immunity in Drosophila melanogaster. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e344. [PMID: 30993906 DOI: 10.1002/wdev.344] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
From flies to humans, many components of the innate immune system have been conserved during metazoan evolution. This foundational observation has allowed us to develop Drosophila melanogaster, the fruit fly, into a powerful model to study innate immunity in animals. Thanks to an ever-growing arsenal of genetic tools, an easily manipulated genome, and its winning disposition, Drosophila is now employed to study not only basic molecular mechanisms of pathogen recognition and immune signaling, but also the nature of physiological responses activated in the host by microbial challenge and how dysregulation of these processes contributes to disease. Here, we present a collection of methods and protocols to challenge the fly with an assortment of microbes, both systemically and orally, and assess its humoral, cellular, and epithelial response to infection. Our review covers techniques for measuring the reaction to microbial infection both qualitatively and quantitatively. Specifically, we describe survival, bacterial load, BLUD (a measure of disease tolerance), phagocytosis, melanization, clotting, and ROS production assays, as well as efficient protocols to collect hemolymph and measure immune gene expression. We also offer an updated catalog of online resources and a collection of popular reporter lines and mutants to facilitate research efforts. This article is categorized under: Technologies > Analysis of Cell, Tissue, and Animal Phenotypes.
Collapse
Affiliation(s)
- Katia Troha
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York
| | - Nicolas Buchon
- Department of Entomology, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York
| |
Collapse
|
50
|
Lee WS, Webster JA, Madzokere ET, Stephenson EB, Herrero LJ. Mosquito antiviral defense mechanisms: a delicate balance between innate immunity and persistent viral infection. Parasit Vectors 2019; 12:165. [PMID: 30975197 PMCID: PMC6460799 DOI: 10.1186/s13071-019-3433-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/04/2019] [Indexed: 01/24/2023] Open
Abstract
Mosquito-borne diseases are associated with major global health burdens. Aedes spp. and Culex spp. are primarily responsible for the transmission of the most medically important mosquito-borne viruses, including dengue virus, West Nile virus and Zika virus. Despite the burden of these pathogens on human populations, the interactions between viruses and their mosquito hosts remain enigmatic. Viruses enter the midgut of a mosquito following the mosquito’s ingestion of a viremic blood meal. During infection, virus recognition by the mosquito host triggers their antiviral defense mechanism. Of these host defenses, activation of the RNAi pathway is the main antiviral mechanism, leading to the degradation of viral RNA, thereby inhibiting viral replication and promoting viral clearance. However, whilst antiviral host defense mechanisms limit viral replication, the mosquito immune system is unable to effectively clear the virus. As such, these viruses can establish persistent infection with little or no fitness cost to the mosquito vector, ensuring life-long transmission to humans. Understanding of the mosquito innate immune response enables the discovery of novel antivectorial strategies to block human transmission. This review provides an updated and concise summary of recent studies on mosquito antiviral immune responses, which is a key determinant for successful virus transmission. In addition, we will also discuss the factors that may contribute to persistent infection in mosquito hosts. Finally, we will discuss current mosquito transmission-blocking strategies that utilize genetically modified mosquitoes and Wolbachia-infected mosquitoes for resistance to pathogens.
Collapse
Affiliation(s)
- Wai-Suet Lee
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Julie A Webster
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Eugene T Madzokere
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Eloise B Stephenson
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia.,Environmental Futures Research Institute, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia.
| |
Collapse
|