1
|
Leelagud P, Wang HL, Lu KH, Dai SM. Pseudomonas mosselii: a potential alternative for managing pyrethroid-resistant Aedes aegypti. PEST MANAGEMENT SCIENCE 2024; 80:4344-4351. [PMID: 38634536 DOI: 10.1002/ps.8139] [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: 12/17/2023] [Revised: 03/23/2024] [Accepted: 04/18/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Aedes aegypti is a widespread mosquito in tropical and subtropical regions that causes significant mortality and morbidity in humans by transmitting diseases, such as dengue fever and Zika virus disease. Synthetic insecticides, such as pyrethroids, have been used to control Ae. aegypti, but these insecticides can also affect nontarget organisms and contaminate soil and water. This study aimed to investigate the mosquitocidal activity of Pseudomonas mosselii isolated from pond sludge against larvae of Ae. aegypti. RESULTS Based on the initial results, similar time-course profiles were obtained for the mosquitocidal activity of the bacterial culture and its supernatant, and the pellet resuspended in Luria-Bertani (LB) medium also showed delayed toxicity. These results imply that the toxic component can be released into the medium from live bacteria. Further research indicated that the toxic component appeared in the supernatant approximately 4 h after a 3-mL stock was cultured in 200 mL of LB medium. The stabilities of the P. mosselii culture and supernatant stored at different temperatures were also evaluated, and the best culture stability was obtained at 28 °C and supernatant stability at 4 °C. The bacterial culture and supernatant were toxic to larvae and pupae of not only susceptible Ae. aegypti but also pyrethroid-resistant strains. CONCLUSION This study highlights the value of the mosquitocidal activity of P. mosselii, which has potential as an alternative insecticide to control pyrethroid-resistant Ae. aegypti in the field. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Piyatida Leelagud
- Department of Entomology, National Chung Hsing University, Taichung, Taiwan
| | - Hui-Liang Wang
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - Kuang-Hui Lu
- Department of Entomology, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Mei Dai
- Department of Entomology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
2
|
McMillan IA, Norris MH, Heacock-Kang Y, Zarzycki-Siek J, Sun Z, Hartney BA, Filipowska LK, Islam MN, Crick DC, Borlee BR, Hoang TT. TetR-like regulator BP1026B_II1561 controls aromatic amino acid biosynthesis and intracellular pathogenesis in Burkholderia pseudomallei. Front Microbiol 2024; 15:1441330. [PMID: 39211319 PMCID: PMC11358695 DOI: 10.3389/fmicb.2024.1441330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Burkholderia pseudomallei (Bp) causes the tropical disease melioidosis that afflicts an estimated 165,000 people each year. Bp is a facultative intracellular pathogen that transits through distinct intracellular stages including attachment to host cells, invasion through the endocytic pathway, escape from the endosome, replication in the cytoplasm, generation of protrusions towards neighboring cells, and host cell fusion allowing Bp infection to spread without exiting the intracellular environment. We have identified a TetR-like transcriptional regulator, BP1026B_II1561, that is up-regulated during the late stages of infection as Bp protrudes toward neighboring cells. We have characterized BP1026B_II1561 and determined that it has a role in pathogenesis. A deletional mutant of BP1026B_II1561 is attenuated in RAW264.7 macrophage and BALB/c mouse models of infection. Using RNA-seq, we found that BP1026B_II1561 controls secondary metabolite biosynthesis, fatty acid degradation, and propanoate metabolism. In addition, we identified that BP1026B_II1561 directly controls expression of an outer membrane porin and genes in the shikimate biosynthetic pathway using ChIP-seq. Transposon mutants of genes within the BP1026B_II1561 regulon show defects during intracellular replication in RAW264.7 cells confirming the role of this transcriptional regulator and the pathways it controls in pathogenesis. BP1026B_II1561 also up-regulates the majority of the enzymes in shikimate and tryptophan biosynthetic pathways, suggesting their importance for Bp physiology. To investigate this, we tested fluorinated analogs of anthranilate and tryptophan, intermediates and products of the shikimate and tryptophan biosynthetic pathways, respectively, and showed inhibition of Bp growth at nanomolar concentrations. The expression of these pathways by BP1026b_II1561 and during intracellular infection combined with the inhibition of Bp growth by fluorotryptophan/anthranilate highlights these pathways as potential targets for therapeutic intervention against melioidosis. In the present study, we have identified BP1026B_II1561 as a critical transcriptional regulator for Bp pathogenesis and partially characterized its role during host cell infection.
Collapse
Affiliation(s)
- Ian A. McMillan
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| | - Michael H. Norris
- Pathogen Analysis and Translational Health Group, School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| | - Yun Heacock-Kang
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| | - Jan Zarzycki-Siek
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| | - Zhenxin Sun
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| | - Brooke A. Hartney
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Liliana K. Filipowska
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - M. Nurul Islam
- Department of Chemistry, Biochemistry, and Physics, South Dakota State University, Brookings, SD, United States
| | - Dean C. Crick
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Bradley R. Borlee
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Tung T. Hoang
- School of Life Sciences, University of Hawaiʻi at Mānoa, Honolulu, HI, United States
| |
Collapse
|
3
|
Paliwal D, Rabiey M, Mauchline TH, Hassani-Pak K, Nauen R, Wagstaff C, Andrews S, Bass C, Jackson RW. Multiple toxins and a protease contribute to the aphid-killing ability of Pseudomonas fluorescens PpR24. Environ Microbiol 2024; 26:e16604. [PMID: 38561900 DOI: 10.1111/1462-2920.16604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
Aphids are globally important pests causing damage to a broad range of crops. Due to insecticide resistance, there is an urgent need to develop alternative control strategies. In our previous work, we found Pseudomonas fluorescens PpR24 can orally infect and kill the insecticide-resistant green-peach aphid (Myzus persicae). However, the genetic basis of the insecticidal capability of PpR24 remains unclear. Genome sequencing of PpR24 confirmed the presence of various insecticidal toxins such as Tc (toxin complexes), Rhs (rearrangement hotspot) elements, and other insect-killing proteases. Upon aphids infection with PpR24, RNA-Seq analysis revealed 193 aphid genes were differentially expressed with down-regulation of 16 detoxification genes. In addition, 1325 PpR24 genes (542 were upregulated and 783 downregulated) were subject to differential expression, including genes responsible for secondary metabolite biosynthesis, the iron-restriction response, oxidative stress resistance, and virulence factors. Single and double deletion of candidate virulence genes encoding a secreted protease (AprX) and four toxin components (two TcA-like; one TcB-like; one TcC-like insecticidal toxins) showed that all five genes contribute significantly to aphid killing, particularly AprX. This comprehensive host-pathogen transcriptomic analysis provides novel insight into the molecular basis of bacteria-mediated aphid mortality and the potential of PpR24 as an effective biocontrol agent.
Collapse
Affiliation(s)
- Deepa Paliwal
- School of Biological Sciences, University of Reading, Reading, UK
| | - Mojgan Rabiey
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Tim H Mauchline
- Sustainable Soils and Crops, Rothamsted Research, Harpenden, UK
| | | | | | - Carol Wagstaff
- School of Chemistry, Food and Pharmacy, University of Reading, Reading, UK
| | - Simon Andrews
- School of Biological Sciences, University of Reading, Reading, UK
| | | | - Robert W Jackson
- School of Biological Sciences, University of Reading, Reading, UK
- School of Biosciences and Birmingham Institute of Forest Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
4
|
Zhou L, Meng G, Zhu L, Ma L, Chen K. Insect Antimicrobial Peptides as Guardians of Immunity and Beyond: A Review. Int J Mol Sci 2024; 25:3835. [PMID: 38612644 PMCID: PMC11011964 DOI: 10.3390/ijms25073835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Antimicrobial peptides (AMPs), as immune effectors synthesized by a variety of organisms, not only constitute a robust defense mechanism against a broad spectrum of pathogens in the host but also show promising applications as effective antimicrobial agents. Notably, insects are significant reservoirs of natural AMPs. However, the complex array of variations in types, quantities, antimicrobial activities, and production pathways of AMPs, as well as evolution of AMPs across insect species, presents a significant challenge for immunity system understanding and AMP applications. This review covers insect AMP discoveries, classification, common properties, and mechanisms of action. Additionally, the types, quantities, and activities of immune-related AMPs in each model insect are also summarized. We conducted the first comprehensive investigation into the diversity, distribution, and evolution of 20 types of AMPs in model insects, employing phylogenetic analysis to describe their evolutionary relationships and shed light on conserved and distinctive AMP families. Furthermore, we summarize the regulatory pathways of AMP production through classical signaling pathways and additional pathways associated with Nitric Oxide, insulin-like signaling, and hormones. This review advances our understanding of AMPs as guardians in insect immunity systems and unlocks a gateway to insect AMP resources, facilitating the use of AMPs to address food safety concerns.
Collapse
Affiliation(s)
- Lizhen Zhou
- Department of Plant Protection, College of Plant Protection, Yangzhou University, Yangzhou 225009, China;
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China
| | - Guanliang Meng
- Zoological Research Museum Alexander Koenig, Leibniz Institute for the Analysis of Biodiversity Change, 53113 Bonn, Germany;
| | - Ling Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China;
| | - Li Ma
- College of Plant Protection, Shanxi Agricultural University, Taigu 030810, China
| | - Kangkang Chen
- Department of Plant Protection, College of Plant Protection, Yangzhou University, Yangzhou 225009, China;
| |
Collapse
|
5
|
Lee JH. Host-Microbe Interactions Regulate Intestinal Stem Cells and Tissue Turnover in Drosophila. Int J Stem Cells 2024; 17:51-58. [PMID: 38123486 PMCID: PMC10899887 DOI: 10.15283/ijsc23172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
With the activity of intestinal stem cells and continuous turnover, the gut epithelium is one of the most dynamic tissues in animals. Due to its simple yet conserved tissue structure and enteric cell composition as well as advanced genetic and histologic techniques, Drosophila serves as a valuable model system for investigating the regulation of intestinal stem cells. The Drosophila gut epithelium is in constant contact with indigenous microbiota and encounters externally introduced "non-self" substances, including foodborne pathogens. Therefore, in addition to its role in digestion and nutrient absorption, another essential function of the gut epithelium is to control the expansion of microbes while maintaining its structural integrity, necessitating a tissue turnover process involving intestinal stem cell activity. As a result, the microbiome and pathogens serve as important factors in regulating intestinal tissue turnover. In this manuscript, I discuss crucial discoveries revealing the interaction between gut microbes and the host's innate immune system, closely associated with the regulation of intestinal stem cell proliferation and differentiation, ultimately contributing to epithelial homeostasis.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- National Creative Research Initiative Center for Hologenomics and School of Biological Sciences, Seoul National University, Seoul, Korea
- The Research Institute of Basic Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
6
|
Xu X, Foley E. Vibrio cholerae arrests intestinal epithelial proliferation through T6SS-dependent activation of the bone morphogenetic protein pathway. Cell Rep 2024; 43:113750. [PMID: 38340318 DOI: 10.1016/j.celrep.2024.113750] [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: 07/05/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
To maintain an effective barrier, intestinal progenitor cells must divide at a rate that matches the loss of dead and dying cells. Otherwise, epithelial breaches expose the host to systemic infection by gut-resident microbes. Unlike most pathogens, Vibrio cholerae blocks tissue repair by arresting progenitor proliferation in the Drosophila model. At present, we do not understand how V. cholerae circumvents such a critical antibacterial defense. We find that V. cholerae blocks epithelial repair by activating the growth inhibitor bone morphogenetic protein (BMP) pathway in progenitors. Specifically, we show that interactions between V. cholerae and gut commensals initiate BMP signaling via host innate immune defenses. Notably, we find that V. cholerae also activates BMP and arrests proliferation in zebrafish intestines, indicating an evolutionarily conserved link between infection and failure in tissue repair. Our study highlights how enteric pathogens engage host immune and growth regulatory pathways to disrupt intestinal epithelial repair.
Collapse
Affiliation(s)
- Xinyue Xu
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
7
|
Sułek M, Kordaczuk J, Mak P, Śmiałek-Bartyzel J, Hułas-Stasiak M, Wojda I. Immune priming modulates Galleria mellonella and Pseudomonas entomophila interaction. Antimicrobial properties of Kazal peptide Pr13a. Front Immunol 2024; 15:1358247. [PMID: 38469316 PMCID: PMC10925678 DOI: 10.3389/fimmu.2024.1358247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/23/2024] [Indexed: 03/13/2024] Open
Abstract
Galleria mellonella larvae repeatedly infected with Pseudomonas entomophila bacteria re-induced their immune response. Its parameters, i.e. the defence activities of cell-free hemolymph, the presence and activity of antimicrobial peptides, and the expression of immune-relevant genes were modulated after the re-challenge in comparison to non-primed infected larvae, resulting in better protection. No enhanced resistance was observed when the larvae were initially infected with other microorganisms, and larvae pre-infected with P. entomophila were not more resistant to further infection with other pathogens. Then, the peptide profiles of hemolymph from primed- and non-primed larvae infected with P. entomophila were compared by quantitative RP-HPLC (Reverse Phase - High Performance Liquid Chromatography). The level of carbonic anhydrase, anionic peptide-1, proline peptide-2, and finally, unknown so far, putative Kazal peptide Pr13a was higher in the primed infected animals than in the larvae infected with P. entomophila for the first time. The expression of the Pr13a gene increased two-fold after the infection, but only in the primed animals. To check whether the enhanced level of Pr13a could have physiological significance, the peptide was purified to homogeneity and checked for its defence properties. In fact, it had antibacterial activity: at the concentration of 15 µM and 7.5 µM it reduced the number of P. entomophila and Bacillus thuringiensis CFU, respectively, to about 40%. The antibacterial activity of Pr13a was correlated with changes observed on the surface of the peptide-treated bacteria, e.g. surface roughness and adhesion force. The presented results bring us closer to finding hemolymph constituents responsible for the effect of priming on the immune response in re-infected insects.
Collapse
Affiliation(s)
- Michał Sułek
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| | - Jakub Kordaczuk
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| | - Paweł Mak
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Justyna Śmiałek-Bartyzel
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| | - Iwona Wojda
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Lublin, Poland
| |
Collapse
|
8
|
Moyano A, Croce AC, Scolari F. Pathogen-Mediated Alterations of Insect Chemical Communication: From Pheromones to Behavior. Pathogens 2023; 12:1350. [PMID: 38003813 PMCID: PMC10675518 DOI: 10.3390/pathogens12111350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Pathogens can influence the physiology and behavior of both animal and plant hosts in a manner that promotes their own transmission and dispersal. Recent research focusing on insects has revealed that these manipulations can extend to the production of pheromones, which are pivotal in chemical communication. This review provides an overview of the current state of research and available data concerning the impacts of bacterial, viral, fungal, and eukaryotic pathogens on chemical communication across different insect orders. While our understanding of the influence of pathogenic bacteria on host chemical profiles is still limited, viral infections have been shown to induce behavioral changes in the host, such as altered pheromone production, olfaction, and locomotion. Entomopathogenic fungi affect host chemical communication by manipulating cuticular hydrocarbons and pheromone production, while various eukaryotic parasites have been observed to influence insect behavior by affecting the production of pheromones and other chemical cues. The effects induced by these infections are explored in the context of the evolutionary advantages they confer to the pathogen. The molecular mechanisms governing the observed pathogen-mediated behavioral changes, as well as the dynamic and mutually influential relationships between the pathogen and its host, are still poorly understood. A deeper comprehension of these mechanisms will prove invaluable in identifying novel targets in the perspective of practical applications aimed at controlling detrimental insect species.
Collapse
Affiliation(s)
- Andrea Moyano
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| | - Anna Cleta Croce
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| | - Francesca Scolari
- Institute of Molecular Genetics, Italian National Research Council (CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy; (A.M.); (A.C.C.)
- Department of Biology and Biotechnology, University of Pavia, Via Ferrata 9, I-27100 Pavia, Italy
| |
Collapse
|
9
|
Grandy S, Scur M, Dolan K, Nickerson R, Cheng Z. Using model systems to unravel host-Pseudomonas aeruginosa interactions. Environ Microbiol 2023; 25:1765-1784. [PMID: 37290773 DOI: 10.1111/1462-2920.16440] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
Using model systems in infection biology has led to the discoveries of many pathogen-encoded virulence factors and critical host immune factors to fight pathogenic infections. Studies of the remarkable Pseudomonas aeruginosa bacterium that infects and causes disease in hosts as divergent as humans and plants afford unique opportunities to shed new light on virulence strategies and host defence mechanisms. One of the rationales for using model systems as a discovery tool to characterise bacterial factors driving human infection outcomes is that many P. aeruginosa virulence factors are required for pathogenesis in diverse different hosts. On the other side, many host signalling components, such as the evolutionarily conserved mitogen-activated protein kinases, are involved in immune signalling in a diverse range of hosts. Some model organisms that have less complex immune systems also allow dissection of the direct impacts of innate immunity on host defence without the interference of adaptive immunity. In this review, we start with discussing the occurrence of P. aeruginosa in the environment and the ability of this bacterium to cause disease in various hosts as a natural opportunistic pathogen. We then summarise the use of some model systems to study host defence and P. aeruginosa virulence.
Collapse
Affiliation(s)
- Shannen Grandy
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michal Scur
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kathleen Dolan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Rhea Nickerson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
10
|
Kordaczuk J, Sułek M, Mak P, Śmiałek-Bartyzel J, Hułas-Stasiak M, Wojda I. Defence response of Galleria mellonella larvae to oral and intrahemocelic infection with Pseudomonasentomophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104749. [PMID: 37279831 DOI: 10.1016/j.dci.2023.104749] [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: 02/01/2023] [Revised: 05/10/2023] [Accepted: 05/29/2023] [Indexed: 06/08/2023]
Abstract
We report differences in the course of infection of G. mellonella larvae with P. entomophila via intrahemocelic and oral routes. Survival curves, larval morphology, histology, and induction of defence response were investigated. Larvae injected with 10 and 50 cells of P. entomophila activated a dose-dependent immune response, which was manifested by induction of immune-related genes and dose-dependent defence activity in larval hemolymph. In contrast, after the oral application of the pathogen, antimicrobial activity was detected in whole hemolymph of larvae infected with the 103 but not 105 dose in spite of the induction of immune response manifested as immune-relevant gene expression and defence activity of electrophoretically separated low-molecular hemolymph components. Among known proteins induced after the P. entomophila infection, we identified proline-rich peptide 1 and 2, cecropin D-like peptide, galiomycin, lysozyme, anionic peptide 1, defensin-like peptide, and a 27 kDa hemolymph protein. The expression of the lysozyme gene and the amount of protein in the hemolymph were correlated with inactivity of hemolymph in insects orally infected with a higher dose of P. entomophila, pointing to its role in the host-pathogen interaction.
Collapse
Affiliation(s)
- Jakub Kordaczuk
- Maria Curie-Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Lublin, Poland
| | - Michał Sułek
- Maria Curie-Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Lublin, Poland
| | - Paweł Mak
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Analytical Biochemistry, Kraków, Poland
| | - Justyna Śmiałek-Bartyzel
- Jagiellonian University, Faculty of Biochemistry, Biophysics and Biotechnology, Department of Analytical Biochemistry, Kraków, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Kraków, Poland
| | - Monika Hułas-Stasiak
- Maria Curie-Sklodowska University, Institute of Biological Sciences, Department of Functional Anatomy and Cytobiology, Lublin, Poland
| | - Iwona Wojda
- Maria Curie-Sklodowska University, Institute of Biological Sciences, Department of Immunobiology, Lublin, Poland.
| |
Collapse
|
11
|
Qush A, Al Khatib HA, Rachid H, Al-Tamimi H, Al-Eshaq A, Al-Adwi S, Yassine HM, Kamareddine L. Intake of caffeine containing sugar diet remodels gut microbiota and perturbs Drosophila melanogaster immunity and lifespan. Microbes Infect 2023; 25:105149. [PMID: 37169244 DOI: 10.1016/j.micinf.2023.105149] [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: 11/30/2022] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
The diet-microbiome-immunity axis is one among the many arms that draw up the "we are what we intake" proclamation. As such, studies on the effect of food and beverage intake on the gut environment and microbiome and on modulating immunological responses and the host's susceptibility to pathogens are on the rise. A typical accompaniment in different sustenance we consume on daily basis is the trimethylxanthine alkaloid caffeine. Being a chief component in our regular aliment, a better understanding of the effect of caffeine containing food and beverages on our gut-microbiome-immunity axis and henceforth on our health is much needed. In this study, we shed more light on the effect of oral consumption of caffeine supplemented sugar diet on the gut environment, specifically on the gut microbiota, innate immunity and host susceptibility to pathogens using the Drosophila melanogaster model organism. Our findings reveal that the oral intake of a dose-specific caffeine containing sucrose/agarose sugar diet causes a significant alteration within the fly gut milieu demarcated by microbial dysbiosis and an elevation in the production of reactive oxygen species and expression of immune-deficiency (Imd) pathway-dependent antimicrobial peptide genes. The oral intake of caffeine containing sucrose/agarose sugar diet also renders the flies more susceptible to bacterial infection and shortens their lifespan in both infection and non-infection settings. Our findings set forth additional insight into the potentiality of diet to alter the gut milieu and highlight the importance of dietary control on health.
Collapse
Affiliation(s)
- Abeer Qush
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hebah A Al Khatib
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Hajar Rachid
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hend Al-Tamimi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Alyaa Al-Eshaq
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Shaima Al-Adwi
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Hadi M Yassine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| | - Layla Kamareddine
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
12
|
Raval D, Daley L, Eleftherianos I. Drosophila melanogaster larvae are tolerant to oral infection with the bacterial pathogen Photorhabdus luminescens. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000938. [PMID: 37711508 PMCID: PMC10498274 DOI: 10.17912/micropub.biology.000938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/10/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
The fruit fly Drosophila melanogaster is an excellent model for dissecting the molecular and functional bases of bacterial pathogenicity and host antibacterial immune response. The Gram-negative bacterium Photorhabdus luminescens is an insect-specific pathogen that forms a mutualistic relationship with the entomopathogenic nematode Heterorhabditis bacteriophora . Here we find that oral infection of D. melanogaster larvae with P. luminescens moderately reduces their survival ability while the bacteria replicate efficiently in the infected insects. This information will contribute towards understanding host gut immunity against potent bacterial pathogens.
Collapse
Affiliation(s)
- Dhaivat Raval
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Lillia Daley
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
13
|
Joshi M, Viallat-Lieutaud A, Royet J. Role of Rab5 early endosomes in regulating Drosophila gut antibacterial response. iScience 2023; 26:107335. [PMID: 37529104 PMCID: PMC10387576 DOI: 10.1016/j.isci.2023.107335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/31/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Interactions between prokaryotes and eukaryotes require a dialogue between MAMPs and PRRs. In Drosophila, bacterial peptidoglycan is detected by PGRP receptors. While the components of the signaling cascades activated upon PGN/PGRP interactions are well characterized, little is known about the subcellular events that translate these early signaling steps into target gene transcription. Using a Drosophila enteric infection model, we show that gut-associated bacteria can induce the formation of intracellular PGRP-LE aggregates which colocalized with the early endosome marker Rab5. Combining microscopic and RNA-seq analysis, we demonstrate that RNAi inactivation of the endocytosis pathway in the Drosophila gut affects the expression of essential regulators of the NF-κB response leading not only to a disruption of the immune response locally in the gut but also at the systemic level. This work sheds new light on the involvement of the endocytosis pathway in the control of the gut response to intestinal bacterial infection.
Collapse
Affiliation(s)
- Manish Joshi
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Annelise Viallat-Lieutaud
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| | - Julien Royet
- Aix-Marseille Université, CNRS, IBDM-UMR7288, Turing Center for Living Systems, 13009 Marseille, France
| |
Collapse
|
14
|
Marzhoseyni Z, Mousavi MJ, Saffari M, Ghotloo S. Immune escape strategies of Pseudomonas aeruginosa to establish chronic infection. Cytokine 2023; 163:156135. [PMID: 36724716 DOI: 10.1016/j.cyto.2023.156135] [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: 08/08/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/02/2023]
Abstract
The infection caused by P. aeruginosa still is dangerous throughout the world. This is partly due to its immune escape mechanisms considerably increasing the bacterial survival in the host. By escape from recognition by TLRs, interference with complement system activation, phagocytosis inhibition, production of ROS, inhibition of NET production, interference with the generation of cytokines, inflammasome inhibition, reduced antigen presentation, interference with cellular and humoral immunity, and induction of apoptotic cell death and MDSc, P. aeruginosa breaks down the barriers of the immune system and causes lethal infections in the host. Recognition of other immune escape mechanisms of P. aeruginosa may provide a basis for the future treatment of the infection. This manuscript may provide new insights and information for the development of new strategies to combat P. aeruginosa infection. In the present manuscript, the escape mechanisms of P. aeruginosa against immune response would be reviewed.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahmood Saffari
- Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ghotloo
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
15
|
Acken KA, Li B. Pseudomonas virulence factor controls expression of virulence genes in Pseudomonas entomophila. PLoS One 2023; 18:e0284907. [PMID: 37200397 DOI: 10.1371/journal.pone.0284907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 04/11/2023] [Indexed: 05/20/2023] Open
Abstract
Quorum sensing is a communication strategy that bacteria use to collectively alter gene expression in response to cell density. Pathogens use quorum sensing systems to control activities vital to infection, such as the production of virulence factors and biofilm formation. The Pseudomonas virulence factor (pvf) gene cluster encodes a signaling system (Pvf) that is present in over 500 strains of proteobacteria, including strains that infect a variety of plant and human hosts. We have shown that Pvf regulates the production of secreted proteins and small molecules in the insect pathogen Pseudomonas entomophila L48. Here, we identified genes that are likely regulated by Pvf using the model strain P. entomophila L48 which does not contain other known quorum sensing systems. Pvf regulated genes were identified through comparing the transcriptomes of wildtype P. entomophila and a pvf deletion mutant (ΔpvfA-D). We found that deletion of pvfA-D affected the expression of approximately 300 genes involved in virulence, the type VI secretion system, siderophore transport, and branched chain amino acid biosynthesis. Additionally, we identified seven putative biosynthetic gene clusters with reduced expression in ΔpvfA-D. Our results indicate that Pvf controls multiple virulence mechanisms in P. entomophila L48. Characterizing genes regulated by Pvf will aid understanding of host-pathogen interactions and development of anti-virulence strategies against P. entomophila and other pvf-containing strains.
Collapse
Affiliation(s)
- Katie A Acken
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Bo Li
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
16
|
Yao Z, Cai Z, Ma Q, Bai S, Wang Y, Zhang P, Guo Q, Gu J, Lemaitre B, Zhang H. Compartmentalized PGRP expression along the dipteran Bactrocera dorsalis gut forms a zone of protection for symbiotic bacteria. Cell Rep 2022; 41:111523. [DOI: 10.1016/j.celrep.2022.111523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/18/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
|
17
|
Savola E, Vale PF, Walling CA. Larval diet affects adult reproduction, but not survival, independent of the effect of injury and infection in Drosophila melanogaster. JOURNAL OF INSECT PHYSIOLOGY 2022; 142:104428. [PMID: 35932926 DOI: 10.1016/j.jinsphys.2022.104428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Early-life conditions have profound effects on many life-history traits, where early-life diet affects both juvenile development, and adult survival and reproduction. Early-life diet also has consequences for the ability of adults to withstand environmental challenges such as starvation, temperature and desiccation. However, it is less well known how early-life diet influences the consequences of infection in adults. Here we test whether varying the larval diet of female Drosophila melanogaster (through altering protein to carbohydrate ratio, P:C) influences the long-term consequences of injury and infection with the bacterial pathogen Pseudomonasentomophila. Given previous work manipulating adult dietary P:C, we predicted that adults from larvae raised on higher P:C diets would have increased reproduction, but shorter lifespans and an increased rate of ageing, and that the lowest larval P:C diets would be particularly detrimental for adult survival in infected individuals. For larval development, we predicted that low P:C would lead to a longer development time and lower viability. We found that early-life and lifetime egg production were highest at intermediate to high larval P:C diets, but this was independent of injury and infection. There was no effect of larval P:C on adult survival. Larval development was quickest on intermediate P:C and egg-to-pupae and egg-to-adult viability were slightly higher on higher P:C. Overall, despite larval P:C affecting several measured traits, we saw no evidence that larval P:C altered the consequence of infection or injury for adult survival or early-life and lifetime reproduction. Taken together, these data suggest that larval diets appear to have a limited impact on the adult life history consequences of infection.
Collapse
Affiliation(s)
- Eevi Savola
- Institute of Evolutionary Biology, School of Biological Sciences, The University of Edinburgh, Ashworth Laboratories, Edinburgh EH9 3FL, UK
| | - Pedro F Vale
- Institute of Evolutionary Biology, School of Biological Sciences, The University of Edinburgh, Ashworth Laboratories, Edinburgh EH9 3FL, UK
| | - Craig A Walling
- Institute of Evolutionary Biology, School of Biological Sciences, The University of Edinburgh, Ashworth Laboratories, Edinburgh EH9 3FL, UK.
| |
Collapse
|
18
|
Decomposing virulence to understand bacterial clearance in persistent infections. Nat Commun 2022; 13:5023. [PMID: 36028497 PMCID: PMC9418333 DOI: 10.1038/s41467-022-32118-1] [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: 10/29/2021] [Accepted: 07/15/2022] [Indexed: 11/08/2022] Open
Abstract
Following an infection, hosts cannot always clear the pathogen, instead either dying or surviving with a persistent infection. Such variation is ecologically and evolutionarily important because it can affect infection prevalence and transmission, and virulence evolution. However, the factors causing variation in infection outcomes, and the relationship between clearance and virulence are not well understood. Here we show that sustained persistent infection and clearance are both possible outcomes across bacterial species showing a range of virulence in Drosophila melanogaster. Variation in virulence arises because of differences in the two components of virulence: bacterial infection intensity inside the host (exploitation), and the amount of damage caused per bacterium (per parasite pathogenicity). As early-phase exploitation increased, clearance rates later in the infection decreased, whereas there was no apparent effect of per parasite pathogenicity on clearance rates. Variation in infection outcomes is thereby determined by how virulence - and its components - relate to the rate of pathogen clearance. Taken together we demonstrate that the virulence decomposition framework is broadly applicable and can provide valuable insights into host-pathogen interactions.
Collapse
|
19
|
Job V, Gomez-Valero L, Renier A, Rusniok C, Bouillot S, Chenal-Francisque V, Gueguen E, Adrait A, Robert-Genthon M, Jeannot K, Panchev P, Elsen S, Fauvarque MO, Couté Y, Buchrieser C, Attrée I. Genomic erosion and horizontal gene transfer shape functional differences of the ExlA toxin in Pseudomonas spp. iScience 2022; 25:104596. [PMID: 35789842 PMCID: PMC9250014 DOI: 10.1016/j.isci.2022.104596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/15/2022] [Accepted: 06/08/2022] [Indexed: 12/31/2022] Open
Abstract
Two-partner secretion (TPS) is widespread in the bacterial world. The pore-forming TPS toxin ExlA of Pseudomonas aeruginosa is conserved in pathogenic and environmental Pseudomonas. While P. chlororaphis and P. entomophila displayed ExlA-dependent killing, P. putida did not cause damage to eukaryotic cells. ExlA proteins interacted with epithelial cell membranes; however, only ExlAPch induced the cleavage of the adhesive molecule E-cadherin. ExlA proteins participated in insecticidal activity toward the larvae of Galleria mellonella and the fly Drosophila melanogaster. Evolutionary analyses demonstrated that the differences in the C-terminal domains are partly due to horizontal movements of the operon within the genus Pseudomonas. Reconstruction of the evolutionary history revealed the complex horizontal acquisitions. Together, our results provide evidence that conserved TPS toxins in environmental Pseudomonas play a role in bacteria-insect interactions and discrete differences in CTDs may determine their specificity and mode of action toward eukaryotic cells. ExlA is a two-partner secreted toxin conserved across Pseudomonas spp. Environmental Pseudomonas strains encode ExlA with different cytotoxic activities ExlA of environmental Pseudomonas strains play a role in bacteria-insect interactions ExlBA operon shows a complex evolutionary history of horizontal gene transfer
Collapse
Affiliation(s)
- Viviana Job
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | - Laura Gomez-Valero
- Institut Pasteur, Université de Paris, CNRS UMR 6047, Unité Biologie des Bactéries Intracellulaires, 75015 Paris, France
| | - Adèle Renier
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | - Christophe Rusniok
- Institut Pasteur, Université de Paris, CNRS UMR 6047, Unité Biologie des Bactéries Intracellulaires, 75015 Paris, France
| | - Stephanie Bouillot
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | - Viviane Chenal-Francisque
- Institut Pasteur, Université de Paris, CNRS UMR 6047, Unité Biologie des Bactéries Intracellulaires, 75015 Paris, France
| | - Erwan Gueguen
- University of Lyon, Université Lyon 1, INSA de Lyon, CNRS UMR 5240 Microbiologie Adaptation et Pathogénie, Lyon, France
| | - Annie Adrait
- Université Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, Grenoble, France
| | - Mylène Robert-Genthon
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | - Katy Jeannot
- Centre National de Référence de la Résistance aux Antibiotiques, Laboratoire de Bactériologie, Centre Hospitalier Universitaire Jean Minjoz, UMR6249 CNRS, Université de Bourgogne-Franche Comté, Besançon, France
| | - Peter Panchev
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | - Sylvie Elsen
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
| | | | - Yohann Couté
- Université Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, Grenoble, France
- CNRS, CEA, FR2048, Grenoble, France
| | - Carmen Buchrieser
- Institut Pasteur, Université de Paris, CNRS UMR 6047, Unité Biologie des Bactéries Intracellulaires, 75015 Paris, France
- Corresponding author
| | - Ina Attrée
- Université Grenoble Alpes, Institute of Structural Biology, Bacterial Pathogenesis and Cellular Responses Team, UMR5075 CNRS, IRIG, CEA, Grenoble, France
- Corresponding author
| |
Collapse
|
20
|
Turner M, Pietri JE. Antimicrobial peptide expression in the cockroach gut during enterobacterial infection is specific and influenced by type III secretion. Biol Open 2022; 11:275513. [PMID: 35611712 PMCID: PMC9167622 DOI: 10.1242/bio.059414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/29/2022] Open
Abstract
Omnivorous synanthropic cockroaches, such as the German cockroach (Blattella germanica), are reservoirs and vectors of enteric bacterial pathogens. A lifestyle conducive to frequent encounters with high loads of diverse bacteria may have led to the evolution of unique innate immune systems in these insects. The innate immune response of insects relies largely on generalized mechanisms to sense and eliminate foreign microbes. However, analyses of the genomes of common synanthropic cockroaches previously revealed a repertoire of pathogen associated molecular pattern (PAMP) receptors and antimicrobial peptides (AMPs) that is significantly expanded relative to most holometabolous insect models and vectors, supporting the intriguing possibility that cockroaches may encode enhanced recognition within their immune system and may possess an enhanced capacity to fine tune innate immune responses. Investigating how cockroaches respond to infection with enterobacteria provides the opportunity to expand our fundamental knowledge of the regulation of insect innate immunity in a context that is biologically and medically relevant. German cockroaches can harbor both Salmonella enterica serovar Typhimurium and Escherichia coli in their gut without experiencing pathogenesis. The former colonizes the gut and replicates while the latter persists only transiently. We hypothesized that differences in the innate immune response may contribute to or result from the difference in infection dynamics between the two enterobacteria. To test this hypothesis, we used qRT-PCR to analyze expression of five genes encoding representative AMPs (Attacins, Blattellicin, Defensins) in the gut of German cockroaches 1 and 24 h after ingestion of live or heat-killed enterobacteria. We found that robust AMP expression was induced in response to ingestion of a live wild-type strain of S. Typhimurium, but not in response to live E. coli, heat-killed S. Typhimurium, or a live mutant strain of S. Typhimurium lacking type III secretion systems. These results indicate that the cockroach immune system does not respond to stimulation with high levels of ingested bacterial PAMPs such as peptidoglycan. Rather, AMP expression in the gut appears to be induced by active bacterial colonization involving type III secretion. We speculate that this form of regulation may have evolved to prevent over activation of the immune system from frequent ingestion of innocuous, non-colonizing, or non-viable bacteria. While additional work is needed to delineate the molecular mechanisms underlying our observations, our findings provide significant novel insight into the immunological adaptation of cockroaches to life in septic environments as well as the factors that regulate bacterial pathogen transmission by these insects.
Collapse
|
21
|
Ahmed S, Sajjadian SM, Kim Y. HMGB1-Like Dorsal Switch Protein 1 Triggers a Damage Signal in Mosquito Gut to Activate Dual Oxidase via Eicosanoids. J Innate Immun 2022; 14:657-672. [PMID: 35512659 PMCID: PMC9801255 DOI: 10.1159/000524561] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/18/2022] [Indexed: 01/03/2023] Open
Abstract
Several mosquitoes transmit human pathogens by blood feeding, with the gut being the main entrance for the pathogens. Thus, the gut epithelium defends the pathogens by eliciting potent immune responses. However, it was unclear how the mosquito gut discriminates pathogens among various microflora in the lumen. This study proposed a hypothesis that a damage signal might be specifically induced by pathogens in the gut. The Asian tiger mosquito, Aedes albopictus, encodes dorsal switch protein 1 (Aa-DSP1) as a putative damage-associated molecular pattern (DAMP). Aa-DSP1 was localized in the nucleus of the midgut epithelium in naïve larvae. Upon infection by a pathogenic bacterium, Serratia marcescens, Aa-DSP1 was released to hemocoel and activated phospholipase A2 (PLA2). The activated PLA2 increased the level of prostaglandin E2 (PGE2) in the gut and subsequently increased Ca2+ signal to produce reactive oxygen species (ROS) via dual oxidase (Duox). Inhibition of Aa-DSP1 via RNA interference or specific inhibitor treatment failed to increase PGE2/Ca2+ signal upon the bacterial infection. Thus, the inhibitors specifically targeting eicosanoid biosynthesis significantly prevented the upregulation of ROS production in the gut and enhanced mosquito mortality after the bacterial infection. However, such inhibitory effects were rescued by adding PGE2. These suggest that Aa-DSP1 plays an important role in immune response of the mosquito gut as a DAMP during pathogen infection by triggering a signaling pathway, DSP1/PLA2/Ca2+/Duox.
Collapse
|
22
|
Armitage SA, Genersch E, McMahon DP, Rafaluk-Mohr C, Rolff J. Tripartite interactions: how immunity, microbiota and pathogens interact and affect pathogen virulence evolution. CURRENT OPINION IN INSECT SCIENCE 2022; 50:100871. [PMID: 34999035 DOI: 10.1016/j.cois.2021.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The bipartite interactions between insect hosts and their bacterial gut microbiota, or their bacterial pathogens, are empirically and theoretically well-explored. However, direct, and indirect tripartite interactions will also likely occur inside a host. These interactions will almost certainly affect the trajectory of pathogen virulence evolution, an area that is currently under researched. The interactions within tripartite associations can be competitive, that is, exploitative-competition, interference-competition or apparent-competition. Competitive interactions will be significantly influenced by non-competitive effects, for example, immunopathology, immunosuppression, and microbiota-mediated tolerance. Considering a combination of these interactions and effects, will enable an increased understanding of the evolution of pathogen virulence. This new perspective allows us to identify several novel research questions, which we hope will be a useful framework for future research.
Collapse
Affiliation(s)
- Sophie Ao Armitage
- Institute of Biology, Freie Universität Berlin, Königin-Luise-Straße 1-3, 14195 Berlin, Germany.
| | - Elke Genersch
- Institute for Bee Research, Friedrich-Engels-Straße 32, 16540 Hohen Neuendorf, Germany; Institute of Microbiology and Epizootics, Faculty of Veterinary Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Dino P McMahon
- Institute of Biology, Freie Universität Berlin, Königin-Luise-Straße 1-3, 14195 Berlin, Germany; Department for Materials and Environment, BAM Federal Institute for Materials Research and Testing, Unter den Eichen 87, 12205 Berlin, Germany
| | - Charlotte Rafaluk-Mohr
- Institute of Biology, Freie Universität Berlin, Königin-Luise-Straße 1-3, 14195 Berlin, Germany
| | - Jens Rolff
- Institute of Biology, Freie Universität Berlin, Königin-Luise-Straße 1-3, 14195 Berlin, Germany; Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
| |
Collapse
|
23
|
Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 2022; 38:110505. [PMID: 35263602 DOI: 10.1016/j.celrep.2022.110505] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key regulator of metabolism and interacts with the intestinal microbiome. Here, we study the role of the Drosophila intestinal stem cell (ISC)-specific biotin transporter Smvt in midgut homeostasis, infection-induced regeneration, and tumorigenesis. We show that Smvt-transported biotin in ISCs is necessary for ISC mitosis. Smvt deficiency impairs intestinal maintenance, which can be rescued by the human Smvt, encoded by SLC5A6. ISC-specific, Smvt-silenced flies exhibit microbial dysbiosis, whereby the growth of Providencia sneebia, an opportunistic pathogen, is favored. Dysbiosis correlates with increased Nox expression, reactive oxygen species (ROS), and enterocyte apoptosis. Flies acquire biotin from their diet and microbiota. We show that, when dietary biotin is scarce, biotin-producing commensals, e.g., E. coli, can rescue reduced ISC mitosis. Smvt and commensals also control intestinal tumor growth. Our findings suggest that direct modification of the gut microbiome by biotin can serve as an approach for the treatment of dysbiosis-promoted diseases and tumorigenesis control.
Collapse
Affiliation(s)
- Constantina Neophytou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus.
| |
Collapse
|
24
|
Soory A, Ratnaparkhi GS. SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 2022; 18:e1010356. [PMID: 35255103 PMCID: PMC8929699 DOI: 10.1371/journal.ppat.1010356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 03/17/2022] [Accepted: 02/09/2022] [Indexed: 12/13/2022] Open
Abstract
Post-translational modification by the small ubiquitin-like modifier, SUMO can modulate the activity of its conjugated proteins in a plethora of cellular contexts. The effect of SUMO conjugation of proteins during an immune response is poorly understood in Drosophila. We have previously identified that the transcription factor Jra, the Drosophila Jun ortholog and a member of the AP-1 complex is one such SUMO target. Here, we find that Jra is a regulator of the Pseudomonas entomophila induced gut immune gene regulatory network, modulating the expression of a few thousand genes, as measured by quantitative RNA sequencing. Decrease in Jra in gut enterocytes is protective, suggesting that reduction of Jra signaling favors the host over the pathogen. In Jra, lysines 29 and 190 are SUMO conjugation targets, with the JraK29R+K190R double mutant being SUMO conjugation resistant (SCR). Interestingly, a JraSCR fly line, generated by CRISPR/Cas9 based genome editing, is more sensitive to infection, with adults showing a weakened host response and increased proliferation of Pseudomonas. Transcriptome analysis of the guts of JraSCR and JraWT flies suggests that lack of SUMOylation of Jra significantly changes core elements of the immune gene regulatory network, which include antimicrobial agents, secreted ligands, feedback regulators, and transcription factors. Mechanistically, SUMOylation attenuates Jra activity, with the TFs, forkhead, anterior open, activating transcription factor 3 and the master immune regulator Relish being important transcriptional targets. Our study implicates Jra as a major immune regulator, with dynamic SUMO conjugation/deconjugation of Jra modulating the kinetics of the gut immune response. The intestine has a resident population of commensal microorganisms against which the immune machinery is tuned to show low or no reactivity. In contrast, when pathogenic microorganisms are ingested, the gut responds by activating signaling cascades that lead to the killing and clearance of the pathogen. In this study, we examine the role played by the well-known transcription factor Jun in regulating the immune response in the Drosophila gut. We find that loss of Jun leads to the change in intensity and kinetics of the gut immune transcriptome. The transcriptional profile indicates a stronger response when Jun activity is reduced. Also, animals infected with Pseudomonas entomophila live longer when Jun signaling is reduced. Further, we find that Jun is post-translationally modified on Lys29 and Lys190 by SUMO. To understand the effect of SUMO-conjugation of Jun, we create by state-of-the-art CRISPR/Cas9 genome editing a Drosophila line where Jun is resistant to SUMOylation. This line is more sensitive to infection, with a weaker host-defense response. Our data suggest that Jun Signaling favors the pathogen by dampening the immune response. SUMO conjugation of Jun reverses the dampening and strengthens the immune response in favor of the host. Dynamic SUMOylation of Jun thus fine-tunes the gut immune response to pathogens.
Collapse
Affiliation(s)
- Amarendranath Soory
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| | - Girish S. Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & Research, Pune, india
- * E-mail: (AS); (GR)
| |
Collapse
|
25
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
26
|
Sina Rahme B, Lestradet M, Di Venanzio G, Ayyaz A, Yamba MW, Lazzaro M, Liégeois S, Garcia Véscovi E, Ferrandon D. The fliR gene contributes to the virulence of S. marcescens in a Drosophila intestinal infection model. Sci Rep 2022; 12:3068. [PMID: 35197500 PMCID: PMC8866479 DOI: 10.1038/s41598-022-06780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/24/2022] [Indexed: 12/05/2022] Open
Abstract
Serratia marcescens is an opportunistic bacterium that infects a wide range of hosts including humans. It is a potent pathogen in a septic injury model of Drosophila melanogaster since a few bacteria directly injected in the body cavity kill the insect within a day. In contrast, flies do not succumb to ingested bacteria for days even though some bacteria cross the intestinal barrier into the hemolymph within hours. The mechanisms by which S. marcescens attacks enterocytes and damages the intestinal epithelium remain uncharacterized. To better understand intestinal infections, we performed a genetic screen for loss of virulence of ingested S. marcescens and identified FliR, a structural component of the flagellum, as a virulence factor. Next, we compared the virulence of two flagellum mutants fliR and flhD in two distinct S. marcescens strains. Both genes are required for S. marcescens to escape the gut lumen into the hemocoel, indicating that the flagellum plays an important role for the passage of bacteria through the intestinal barrier. Unexpectedly, fliR but not flhD is involved in S. marcescens-mediated damages of the intestinal epithelium that ultimately contribute to the demise of the host. Our results therefore suggest a flagellum-independent role for fliR in bacterial virulence.
Collapse
Affiliation(s)
- Bechara Sina Rahme
- Université de Strasbourg, Strasbourg, France
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France
| | - Matthieu Lestradet
- Université de Strasbourg, Strasbourg, France
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France
| | - Gisela Di Venanzio
- Instituto de Biología Molecular y Cellular de Rosario, Consejo Nacional de Investigaciones Cientificas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Arshad Ayyaz
- Université de Strasbourg, Strasbourg, France
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Miriam Wennida Yamba
- Université de Strasbourg, Strasbourg, France
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France
| | - Martina Lazzaro
- Instituto de Biología Molecular y Cellular de Rosario, Consejo Nacional de Investigaciones Cientificas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Samuel Liégeois
- Université de Strasbourg, Strasbourg, France
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France
| | - Eleonora Garcia Véscovi
- Instituto de Biología Molecular y Cellular de Rosario, Consejo Nacional de Investigaciones Cientificas y Tecnológicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Dominique Ferrandon
- Université de Strasbourg, Strasbourg, France.
- UPR 9022 du CNRS, Institut de Biologie Moléculaire du CNRS, CNRS, Strasbourg, France.
| |
Collapse
|
27
|
Sharda S, Kawecki TJ, Hollis B. Adaptation to a bacterial pathogen in Drosophila melanogaster is not aided by sexual selection. Ecol Evol 2022; 12:e8543. [PMID: 35169448 PMCID: PMC8840902 DOI: 10.1002/ece3.8543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 11/08/2022] Open
Abstract
Theory predicts that sexual selection should aid adaptation to novel environments, but empirical support for this idea is limited. Pathogens are a major driver of host evolution and, unlike abiotic selection pressures, undergo epidemiological and co-evolutionary cycles with the host involving adaptation and counteradaptation. Because of this, populations harbor ample genetic variation underlying immunity and the opportunity for sexual selection based on condition-dependent "good genes" is expected to be large. In this study, we evolved populations of Drosophila melanogaster in a 2-way factorial design manipulating sexual selection and pathogen presence, using a gram-negative insect pathogen Pseudomonas entomophila, for 14 generations. We then examined how the presence of sexual selection and the pathogen, as well as any potential interaction, affected the evolution of pathogen resistance. We found increased resistance to P. entomophila in populations that evolved under pathogen pressure, driven primarily by increased female survival after infection despite selection for resistance acting only on males over the course of experimental evolution. This result suggests that the genetic basis of resistance is in part shared between the sexes. We did not find any evidence of sexual selection aiding adaptation to pathogen, however, a finding contrary to the predictions of "good genes" theory. Our results therefore provide no support for a role for sexual selection in the evolution of immunity in this experimental system.
Collapse
Affiliation(s)
- Sakshi Sharda
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Tadeusz J. Kawecki
- Department of Ecology and EvolutionUniversity of LausanneLausanneSwitzerland
| | - Brian Hollis
- Department of Biological SciencesUniversity of South CarolinaColumbiaSouth CarolinaUSA
| |
Collapse
|
28
|
Bahuguna S, Atilano M, Glittenberg M, Lee D, Arora S, Wang L, Zhou J, Redhai S, Boutros M, Ligoxygakis P. Bacterial recognition by PGRP-SA and downstream signalling by Toll/DIF sustain commensal gut bacteria in Drosophila. PLoS Genet 2022; 18:e1009992. [PMID: 35007276 PMCID: PMC8782595 DOI: 10.1371/journal.pgen.1009992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 01/21/2022] [Accepted: 12/14/2021] [Indexed: 11/19/2022] Open
Abstract
The gut sets the immune and metabolic parameters for the survival of commensal bacteria. We report that in Drosophila, deficiency in bacterial recognition upstream of Toll/NF-κB signalling resulted in reduced density and diversity of gut bacteria. Translational regulation factor 4E-BP, a transcriptional target of Toll/NF-κB, mediated this host-bacteriome interaction. In healthy flies, Toll activated 4E-BP, which enabled fat catabolism, which resulted in sustaining of the bacteriome. The presence of gut bacteria kept Toll signalling activity thus ensuring the feedback loop of their own preservation. When Toll activity was absent, TOR-mediated suppression of 4E-BP made fat resources inaccessible and this correlated with loss of intestinal bacterial density. This could be overcome by genetic or pharmacological inhibition of TOR, which restored bacterial density. Our results give insights into how an animal integrates immune sensing and metabolism to maintain indigenous bacteria in a healthy gut.
Collapse
Affiliation(s)
- Shivohum Bahuguna
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Magda Atilano
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Marcus Glittenberg
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Dohun Lee
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Srishti Arora
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Lihui Wang
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Jun Zhou
- German Cancer Research Centre (DKFZ), Division Signalling and Functional Genomics, BioQuant and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Siamak Redhai
- German Cancer Research Centre (DKFZ), Division Signalling and Functional Genomics, BioQuant and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Centre (DKFZ), Division Signalling and Functional Genomics, BioQuant and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Petros Ligoxygakis
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Zhao X, Karpac J. Glutamate metabolism directs energetic trade-offs to shape host-pathogen susceptibility in Drosophila. Cell Metab 2021; 33:2428-2444.e8. [PMID: 34710355 PMCID: PMC9153082 DOI: 10.1016/j.cmet.2021.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/26/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022]
Abstract
Individual hosts within populations often show inter-individual variation in their susceptibility to bacterial pathogen-related diseases. Utilizing Drosophila, we highlight that phenotypic variation in host-pathogen susceptibility within populations is driven by energetic trade-offs, facilitated by infection-mediated changes in glutamate metabolism. Furthermore, host-pathogen susceptibility is conditioned by life history, which adjusts immunometabolic sensing in muscles to direct vitamin-dependent reallocation of host energy substrates from the adipose tissue (i.e., a muscle-adipose tissue axis). Life history conditions inter-individual variation in the activation strength of intra-muscular NF-κB signaling. Limited intra-muscular NF-κB signaling activity allows for enhanced infection-mediated mitochondrial biogenesis and function, which stimulates glutamate dehydrogenase-dependent synthesis of glutamate. Muscle-derived glutamate acts as a systemic metabolite to promote lipid mobilization through modulating vitamin B enzymatic cofactor transport and function in the adipose tissue. This energy substrate reallocation improves pathogen clearance and boosts host survival. Finally, life history events that adjust energetic trade-offs can shape inter-individual variation in host-pathogen susceptibility after infection.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Molecular and Cellular Medicine, Texas A&M University, College of Medicine, Bryan, TX 77807, USA
| | - Jason Karpac
- Department of Molecular and Cellular Medicine, Texas A&M University, College of Medicine, Bryan, TX 77807, USA.
| |
Collapse
|
30
|
Abstract
The gut microbiota affects the physiology and metabolism of animals and its alteration can lead to diseases such as gut dysplasia or metabolic disorders. Several reports have shown that the immune system plays an important role in shaping both bacterial community composition and abundance in Drosophila, and that immune deficit, especially during aging, negatively affects microbiota richness and diversity. However, there has been little study at the effector level to demonstrate how immune pathways regulate the microbiota. A key set of Drosophila immune effectors are the antimicrobial peptides (AMPs), which confer defense upon systemic infection. AMPs and lysozymes, a group of digestive enzymes with antimicrobial properties, are expressed in the gut and are good candidates for microbiota regulation. Here, we take advantage of the model organism Drosophila melanogaster to investigate the role of AMPs and lysozymes in regulation of gut microbiota structure and diversity. Using flies lacking AMPs and newly generated lysozyme mutants, we colonized gnotobiotic flies with a defined set of commensal bacteria and analyzed changes in microbiota composition and abundance in vertical transmission and aging contexts through 16S rRNA gene amplicon sequencing. Our study shows that AMPs and, to a lesser extent, lysozymes are necessary to regulate the total and relative abundance of bacteria in the gut microbiota. We also decouple the direct function of AMPs from the immune deficiency (IMD) signaling pathway that regulates AMPs but also many other processes, more narrowly defining the role of these effectors in the microbial dysbiosis observed in IMD-deficient flies upon aging.
Collapse
|
31
|
Yamashita K, Oi A, Kosakamoto H, Yamauchi T, Kadoguchi H, Kuraishi T, Miura M, Obata F. Activation of innate immunity during development induces unresolved dysbiotic inflammatory gut and shortens lifespan. Dis Model Mech 2021; 14:271978. [PMID: 34448472 PMCID: PMC8405880 DOI: 10.1242/dmm.049103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/16/2022] Open
Abstract
Early-life inflammatory response is associated with risks of age-related pathologies. How transient immune signalling activity during animal development influences life-long fitness is not well understood. Using Drosophila as a model, we find that activation of innate immune pathway IMD signalling in the developing larvae increases adult starvation resistance, decreases food intake, and shortens organismal lifespan. Interestingly, lifespan is shortened by the IMD activation in the larval gut and fat body, while starvation resistance and food intake are altered by that in neurons. The adult flies developed with IMD activation show sustained IMD activity in the gut, despite complete tissue renewal during metamorphosis. The larval IMD activation increases an immuno-stimulative bacterial species Gluconobacter sp. in the gut microbiome, and this dysbiosis is persistent to adulthood. Removing gut microbiota by antibiotics in adult mitigates intestinal immune activation and rescues the shortened lifespan. This study demonstrates that early-life immune activation triggers long-term physiological changes as highlighted as an irreversible gut microbiota alteration, prolonged inflammatory intestine, and concomitant shortening of the organismal lifespan.
Collapse
Affiliation(s)
- Kyoko Yamashita
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ayano Oi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.,Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.,Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Toshitaka Yamauchi
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Hibiki Kadoguchi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Shizenken, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Takayuki Kuraishi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Shizenken, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Fumiaki Obata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.,Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan.,Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
32
|
Botwright NA, Mohamed AR, Slinger J, Lima PC, Wynne JW. Host-Parasite Interaction of Atlantic salmon ( Salmo salar) and the Ectoparasite Neoparamoeba perurans in Amoebic Gill Disease. Front Immunol 2021; 12:672700. [PMID: 34135900 PMCID: PMC8202022 DOI: 10.3389/fimmu.2021.672700] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
Marine farmed Atlantic salmon (Salmo salar) are susceptible to recurrent amoebic gill disease (AGD) caused by the ectoparasite Neoparamoeba perurans over the growout production cycle. The parasite elicits a highly localized response within the gill epithelium resulting in multifocal mucoid patches at the site of parasite attachment. This host-parasite response drives a complex immune reaction, which remains poorly understood. To generate a model for host-parasite interaction during pathogenesis of AGD in Atlantic salmon the local (gill) and systemic transcriptomic response in the host, and the parasite during AGD pathogenesis was explored. A dual RNA-seq approach together with differential gene expression and system-wide statistical analyses of gene and transcription factor networks was employed. A multi-tissue transcriptomic data set was generated from the gill (including both lesioned and non-lesioned tissue), head kidney and spleen tissues naïve and AGD-affected Atlantic salmon sourced from an in vivo AGD challenge trial. Differential gene expression of the salmon host indicates local and systemic upregulation of defense and immune responses. Two transcription factors, znfOZF-like and znf70-like, and their associated gene networks significantly altered with disease state. The majority of genes in these networks are candidates for mediators of the immune response, cellular proliferation and invasion. These include Aurora kinase B-like, rho guanine nucleotide exchange factor 25-like and protein NDNF-like inhibited. Analysis of the N. perurans transcriptome during AGD pathology compared to in vitro cultured N. perurans trophozoites, as a proxy for wild type trophozoites, identified multiple gene candidates for virulence and indicates a potential master regulatory gene system analogous to the two-component PhoP/Q system. Candidate genes identified are associated with invasion of host tissue, evasion of host defense mechanisms and formation of the mucoid lesion. We generated a novel model for host-parasite interaction during AGD pathogenesis through integration of host and parasite functional profiles. Collectively, this dual transcriptomic study provides novel molecular insights into the pathology of AGD and provides alternative theories for future research in a step towards improved management of AGD.
Collapse
Affiliation(s)
- Natasha A Botwright
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Amin R Mohamed
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - Joel Slinger
- Livestock and Aquaculture, CSIRO Agriculture and Food, Woorim, QLD, Australia
| | - Paula C Lima
- Livestock and Aquaculture, CSIRO Agriculture and Food, St Lucia, QLD, Australia
| | - James W Wynne
- Livestock and Aquaculture, CSIRO Agriculture and Food, Hobart, TAS, Australia
| |
Collapse
|
33
|
Zhang X, Huang H, Wu B, Xie J, Viljoen A, Wang W, Mostert D, Xie Y, Fu G, Xiang D, Lyu S, Liu S, Li C. The M35 Metalloprotease Effector FocM35_1 Is Required for Full Virulence of Fusarium oxysporum f. sp. cubense Tropical Race 4. Pathogens 2021; 10:670. [PMID: 34072465 PMCID: PMC8226822 DOI: 10.3390/pathogens10060670] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
Fusarium oxysporum f. sp. cubense tropical race 4 (Foc TR4) causes Fusarium wilt of banana, the most devastating disease on a banana plant. The genome of Foc TR4 encodes many candidate effector proteins. However, little is known about the functions of these effector proteins on their contributions to disease development and Foc TR4 virulence. Here, we discovered a secreted metalloprotease, FocM35_1, which is an essential virulence effector of Foc TR4. FocM35_1 was highly upregulated during the early stages of Foc TR4 infection progress in bananas. The FocM35_1 knockout mutant compromised the virulence of Foc TR4. FocM35_1 could interact with the banana chitinase MaChiA, and it decreased banana chitinase activity. FocM35_1 induced cell death in Nicotiana benthamiana while suppressing the INF1-induced hypersensitive response (HR), and its predicted enzymatic site was required for lesion formation and the suppression to INF1-induced HR on N. benthamiana leaves. Importantly, treatment of banana leaves with recombinant FocM35_1 accelerates Foc TR4 infection. Collectively, our study provides evidence that metalloprotease effector FocM35 seems to contribute to pathogen virulence by inhibiting the host immunity.
Collapse
Affiliation(s)
- Xiaoxia Zhang
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, China;
| | - Huoqing Huang
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| | - Bangting Wu
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| | - Jianghui Xie
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (J.X.); (W.W.)
| | - Altus Viljoen
- Department of Plant Pathology, University of Stellenbosch, Matieland 7602, South Africa; (A.V.); (D.M.)
| | - Wei Wang
- Key Laboratory of Biology and Genetic Resources of Tropical Crops, Ministry of Agriculture, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China; (J.X.); (W.W.)
| | - Diane Mostert
- Department of Plant Pathology, University of Stellenbosch, Matieland 7602, South Africa; (A.V.); (D.M.)
| | - Yanling Xie
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| | - Gang Fu
- Institute of Plant Protection, Guangxi Academy of Agricultural Sciences, Guangxi Key Laboratory of Biology for Crop Diseases and Insect Pests, Nanning 530007, China;
| | - Dandan Xiang
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| | - Shuxia Lyu
- College of Bioscience and Biotechnology, Shenyang Agricultural University, Shenyang 110866, China;
| | - Siwen Liu
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| | - Chunyu Li
- Key Laboratory of South Subtropical Fruit Biology and Genetic Resource Utilization, Ministry of Agriculture, Key Laboratory of Tropical and Subtropical Fruit Tree Research of Guangdong Province, Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (H.H.); (B.W.); (Y.X.); (D.X.); (C.L.)
| |
Collapse
|
34
|
Bai S, Yao Z, Raza MF, Cai Z, Zhang H. Regulatory mechanisms of microbial homeostasis in insect gut. INSECT SCIENCE 2021; 28:286-301. [PMID: 32888254 DOI: 10.1111/1744-7917.12868] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Insects live in incredibly complex environments. The intestinal epithelium of insects is in constant contact with microorganisms, some of which are beneficial and some harmful to the host. Insect gut health and function are maintained through multidimensional mechanisms that can proficiently remove foreign pathogenic microorganisms while effectively maintaining local symbiotic microbial homeostasis. The basic immune mechanisms of the insect gut, such as the dual oxidase-reactive oxygen species (Duox-ROS) system and the immune deficiency (Imd)-signaling pathway, are involved in the maintenance of microbial homeostasis. This paper reviews the role of physical defenses, the Duox-ROS and Imd signaling pathways, the Janus kinase/signal transducers and activators of transcription signaling pathway, and intestinal symbiotic flora in the homeostatic maintenance of the insect gut microbiome.
Collapse
Affiliation(s)
- Shuai Bai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhichao Yao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Fahim Raza
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhaohui Cai
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
35
|
Kretsch AM, Morgan GL, Acken KA, Barr SA, Li B. Pseudomonas Virulence Factor Pathway Synthesizes Autoinducers That Regulate the Secretome of a Pathogen. ACS Chem Biol 2021; 16:501-509. [PMID: 33595276 DOI: 10.1021/acschembio.0c00901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cell-to-cell communication via chemical signals is an essential mechanism that pathogenic bacteria use to coordinate group behaviors and promote virulence. The Pseudomonas virulence factor (pvf) gene cluster is distributed in more than 500 strains of proteobacteria including both plant and human pathogens. The pvf cluster has been implicated in the production of signaling molecules important for virulence; however, the regulatory impact of these signaling molecules on virulence had not been elucidated. Using the insect pathogen Pseudomonas entomophila L48 as a model, we demonstrated that pvf-encoded biosynthetic enzymes produce PVF autoinducers that regulate the expression of pvf genes and a gene encoding the toxin monalysin via quorum sensing. In addition, PVF autoinducers regulate the expression of nearly 200 secreted and membrane proteins, including toxins, motility proteins, and components of the type VI secretion system, which play key roles in bacterial virulence, colonization, and competition with other microbes. Deletion of pvf also altered the secondary metabolome. Six major compounds upregulated by PVF autoinducers were isolated and structurally characterized, including three insecticidal 3-indolyl oxazoles, the labradorins, and three antimicrobial pyrrolizidine alkaloids, the pyreudiones. The signaling properties of PVF autoinducers and their wide-ranging regulatory effects indicate multifaceted roles of PVF in controlling cell physiology and promoting virulence. The broad genome distribution of pvf suggests that PVF-mediated signaling is relevant to many bacteria of agricultural and biomedical significance.
Collapse
|
36
|
Comparative response of Spodoptera litura challenged per os with Serratia marcescens strains differing in virulence. J Invertebr Pathol 2021; 183:107562. [PMID: 33652013 DOI: 10.1016/j.jip.2021.107562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 01/06/2023]
Abstract
Host plays an important role in influencing virulence of a pathogen and efficacy of a biopesticide. The present study was aimed to characterize the possible factors present in Spodoptera litura that influenced pathogenecity of orally ingested S. marcescens strains, differing in their virulence. Fifth instar larvae of S. litura responded differently as challenged by two Serratia marcescens strains, SEN (virulent strain, LC50 7.02 103 cfu/ml) and ICC-4 (non-virulent strain, LC50 1.19 1012 cfu/ml). Considerable increase in activity of lytic enzymes protease and phospholipase was recorded in the gut and hemolymph of larvae fed on diet supplemented with S. marcescens strain ICC-4 as compared to the larvae treated with S. marcescens strain SEN. However, a significant up-regulation of antioxidative enzymes SOD (in foregut and midgut), CAT (in the midgut) and GST (in the foregut and hemolymph) was recorded in larvae fed on diet treated with the virulent S. marcescens strain SEN in comparison to larvae fed on diet treated with the non-virulent S. marcescens strain ICC-4. Activity of defense related enzymes lysozyme and phenoloxidase activity were also higher in the hemolymph of larvae fed with diet treated with S. marcescens strain SEN as compared to hemolymph of S. marcescens strain ICC-4 treated larvae. More number of over-expressed proteins was observed in the gut and hemolymph of S. marcescens strains ICC-4 and SEN treated larvae, respectively. Identification of the selected differentially expressed proteins indicated induction of proteins involved in insect innate immune response (Immunoglobulin I-set domain, Apolipophorin III, leucine rich repeat and Titin) in S. marcescens strain SEN treated larvae. Over-expression of two proteins, actin related protein and mt DNA helicase, were noted in S. marcescens treated larvae with very high levels observed in the non-virulent strain. Up-regulation of homeobox protein was noted only in S. marcescens strain ICC-4 challenged larvae. This study indicated that ingestion of non-virulent S. marcescens strain ICC-4 induced strong immune response in insect gut while there was weak response to the virulent S. marcescens strain SEN which probably resulted in difference in their virulence.
Collapse
|
37
|
Teoh MC, Furusawa G, Veera Singham G. Multifaceted interactions between the pseudomonads and insects: mechanisms and prospects. Arch Microbiol 2021; 203:1891-1915. [PMID: 33634321 DOI: 10.1007/s00203-021-02230-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/19/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Insects and bacteria are the most widespread groups of organisms found in nearly all habitats on earth, establishing diverse interactions that encompass the entire range of possible symbiotic associations from strict parasitism to obligate mutualism. The complexity of their interactions is instrumental in shaping the roles of insects in the environment, meanwhile ensuring the survival and persistence of the associated bacteria. This review aims to provide detailed insight on the multifaceted symbiosis between one of the most versatile bacterial genera, Pseudomonas (Gammaproteobacteria: Pseudomonadaceae) and a diverse group of insect species. The Pseudomonas engages with varied interactions with insects, being either a pathogen or beneficial endosymbiont, as well as using insects as vectors. In addition, this review also provides updates on existing and potential applications of Pseudomonas and their numerous insecticidal metabolites as biocontrol agents against pest insects for the improvement of integrated pest management strategies. Here, we have summarized several known modes of action and the virulence factors of entomopathogenic Pseudomonas strains essential for their pathogenicity against insects. Meanwhile, the beneficial interactions between pseudomonads and insects are currently limited to a few known insect taxa, despite numerous studies reporting identification of pseudomonads in the guts and haemocoel of various insect species. The vector-symbiont association between pseudomonads and insects can be diverse from strict phoresy to a role switch from commensalism to parasitism following a dose-dependent response. Overall, the pseudomonads appeared to have evolved independently to be either exclusively pathogenic or beneficial towards insects.
Collapse
Affiliation(s)
- Miao-Ching Teoh
- Centre for Chemical Biology, Universiti Sains Malaysia, 11900, Bayan Lepas, Penang, Malaysia
| | - Go Furusawa
- Centre for Chemical Biology, Universiti Sains Malaysia, 11900, Bayan Lepas, Penang, Malaysia
| | - G Veera Singham
- Centre for Chemical Biology, Universiti Sains Malaysia, 11900, Bayan Lepas, Penang, Malaysia.
| |
Collapse
|
38
|
Huang A, Lu M, Ling E, Li P, Wang C. A M35 family metalloprotease is required for fungal virulence against insects by inactivating host prophenoloxidases and beyond. Virulence 2021; 11:222-237. [PMID: 32079481 PMCID: PMC7051145 DOI: 10.1080/21505594.2020.1731126] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A diverse family of metalloproteases (MPs) is distributed in eukaryotes. However, the functions of MPs are still understudied. We report that seven MPs belonging to the M35 family are encoded in the genome of the insect pathogenic fungus Metarhizium robertsii. By gene deletions and insect bioassays, we found that one of the M35-family MPs, i.e. MrM35-4, is required for fungal virulence against insect hosts. MrM35-4 is a secretable enzyme and shows a proteolytic activity implicated in facilitating fungal penetration of insect cuticles. After gene rescue and overexpression, insect bioassays indicated that MrM35-4 contributes to inhibiting insect cuticular and hemocyte melanization activities. Enzymatic cleavage assays revealed that the recombinant prophenoloxidases PPO1 and PPO2 of Drosophila melanogaster could be clipped by MrM35-4 in a manner differing from a serine protease that can activate PPO activities. In addition, it was found that MrM35-4 is involved in suppressing antifungal gene expression in insects. Consistent with the evident apoptogenic effect of MrM35-4 on host cells, we found that the PPO mutant flies differentially succumbed to the infections of the wild-type and mutant strains of M. robertsii. Thus, MrM35-4 plays a multifaceted role beyond targeting PPOs during fungus-insect interactions, which represents a previously unsuspected strategy employed by Metarhizium to outmaneuver insect immune defenses.
Collapse
Affiliation(s)
- Antian Huang
- School of Life Science and Technology, Tongji University, Shanghai, China.,CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Mengting Lu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Erjun Ling
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Ping Li
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chengshu Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
39
|
Kobler JM, Rodriguez Jimenez FJ, Petcu I, Grunwald Kadow IC. Immune Receptor Signaling and the Mushroom Body Mediate Post-ingestion Pathogen Avoidance. Curr Biol 2020; 30:4693-4709.e3. [DOI: 10.1016/j.cub.2020.09.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/31/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
|
40
|
Wang B, Yao Y, Wei P, Song C, Wan S, Yang S, Zhu GM, Liu HM. Housefly Phormicin inhibits Staphylococcus aureus and MRSA by disrupting biofilm formation and altering gene expression in vitro and in vivo. Int J Biol Macromol 2020; 167:1424-1434. [PMID: 33202277 DOI: 10.1016/j.ijbiomac.2020.11.096] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/24/2022]
Abstract
The increasing drug resistance of pathogenic bacteria is a crisis that threatens public health. Antimicrobial peptides (AMPs) have been suggested to be potentially effective alternatives to solve this problem. Here, we tested housefly Phormicin-derived peptides for effects on Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA) infections in vitro and in vivo. A decreased bacterial load of MRSA was observed in the mouse scald model after treatment with Phormicin and in the positive control group (vancomycin). A mouse scrape model indicated that Phormicin helps the host fight drug-resistant MRSA infections. The protective effect of Phormicin on MRSA was confirmed in the Hermetia illucens larvae model. Phormicin also disrupted the formation of S. aureus and MRSA biofilms. Furthermore, this effect coincided with the downregulation of biofilm formation-related gene expression (agrC, sigB, RNAIII, altA, rbf, hla, hld, geh and psmɑ). Notably, virulence genes and several regulatory factors were also altered by Phormicin treatment. Based on these findings, housefly Phormicin helps the host inhibit MRSA infection through effects on biofilm formation and related gene networks. Therefore, housefly Phormicin potential represents a candidate agent for clinical MRSA chemotherapy.
Collapse
Affiliation(s)
- Bing Wang
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (Guizhou Medical University), Guiyang 550025, Guizhou, China.
| | - Yang Yao
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - PengWei Wei
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - ChaoRong Song
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Shan Wan
- Department of Microbial Immunology, The first affiliated hospital of Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - SuWen Yang
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China
| | - Gui Ming Zhu
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (Guizhou Medical University), Guiyang 550025, Guizhou, China
| | - Hong Mei Liu
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang 550025, Guizhou, China; Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China; School of Biology and Engineering, Guizhou Medical University, Guiyang 550025, Guizhou, China.
| |
Collapse
|
41
|
Bjedov I, Cochemé HM, Foley A, Wieser D, Woodling NS, Castillo-Quan JI, Norvaisas P, Lujan C, Regan JC, Toivonen JM, Murphy MP, Thornton J, Kinghorn KJ, Neufeld TP, Cabreiro F, Partridge L. Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in Drosophila. PLoS Genet 2020; 16:e1009083. [PMID: 33253201 PMCID: PMC7738165 DOI: 10.1371/journal.pgen.1009083] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 12/15/2020] [Accepted: 08/26/2020] [Indexed: 01/26/2023] Open
Abstract
Increased cellular degradation by autophagy is a feature of many interventions that delay ageing. We report here that increased autophagy is necessary for reduced insulin-like signalling (IIS) to extend lifespan in Drosophila and is sufficient on its own to increase lifespan. We first established that the well-characterised lifespan extension associated with deletion of the insulin receptor substrate chico was completely abrogated by downregulation of the essential autophagy gene Atg5. We next directly induced autophagy by over-expressing the major autophagy kinase Atg1 and found that a mild increase in autophagy extended lifespan. Interestingly, strong Atg1 up-regulation was detrimental to lifespan. Transcriptomic and metabolomic approaches identified specific signatures mediated by varying levels of autophagy in flies. Transcriptional upregulation of mitochondrial-related genes was the signature most specifically associated with mild Atg1 upregulation and extended lifespan, whereas short-lived flies, possessing strong Atg1 overexpression, showed reduced mitochondrial metabolism and up-regulated immune system pathways. Increased proteasomal activity and reduced triacylglycerol levels were features shared by both moderate and high Atg1 overexpression conditions. These contrasting effects of autophagy on ageing and differential metabolic profiles highlight the importance of fine-tuning autophagy levels to achieve optimal healthspan and disease prevention.
Collapse
Affiliation(s)
- Ivana Bjedov
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Helena M. Cochemé
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Andrea Foley
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Daniela Wieser
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Nathaniel S. Woodling
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Jorge Iván Castillo-Quan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston MA, United States of America
- Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston MA, United States of America
| | - Povilas Norvaisas
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Celia Lujan
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Janne M. Toivonen
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- LAGENBIO, Facultad de Veterinaria-IIS, IA2-CITA, CIBERNED, Universidad de Zaragoza, Zaragoza, Spain
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, the Keith Peters Building, University of Cambridge, Cambridge, United Kingdom
| | - Janet Thornton
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kerri J. Kinghorn
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Thomas P. Neufeld
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church St. SE, Minneapolis, MN, United States of America
| | - Filipe Cabreiro
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| |
Collapse
|
42
|
Arora S, Ligoxygakis P. Beyond Host Defense: Deregulation of Drosophila Immunity and Age-Dependent Neurodegeneration. Front Immunol 2020; 11:1574. [PMID: 32774336 PMCID: PMC7387716 DOI: 10.3389/fimmu.2020.01574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Age-dependent neurodegenerative disorders are a set of diseases that affect millions of individuals worldwide. Apart from a small subset that are the result of well-defined inherited autosomal dominant gene mutations (e.g., those encoding the β-amyloid precursor protein and presenilins), our understanding of the genetic network that underscores their pathology, remains scarce. Genome-wide association studies (GWAS) especially in Alzheimer's disease patients and research in Parkinson's disease have implicated inflammation and the innate immune response as risk factors. However, even if GWAS etiology points toward innate immunity, untangling cause, and consequence is a challenging task. Specifically, it is not clear whether predisposition to de-regulated immunity causes an inadequate response to protein aggregation (such as amyloid or α-synuclein) or is the direct cause of this aggregation. Given the evolutionary conservation of the innate immune response in Drosophila and humans, unraveling whether hyperactive immune response in glia have a protective or pathological role in the brain could be a potential strategy in combating age-related neurological diseases.
Collapse
Affiliation(s)
- Srishti Arora
- Laboratory of Cell Biology, Development and Genetics, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Petros Ligoxygakis
- Laboratory of Cell Biology, Development and Genetics, Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Drosophila as a model for studying cystic fibrosis pathophysiology of the gastrointestinal system. Proc Natl Acad Sci U S A 2020; 117:10357-10367. [PMID: 32345720 DOI: 10.1073/pnas.1913127117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is a recessive disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. The most common symptoms include progressive lung disease and chronic digestive conditions. CF is the first human genetic disease to benefit from having five different species of animal models. Despite the phenotypic differences among the animal models and human CF, these models have provided invaluable insight into understanding disease mechanisms at the organ-system level. Here, we identify a member of the ABCC4 family, CG5789, that has the structural and functional properties expected for encoding the Drosophila equivalent of human CFTR, and thus refer to it as Drosophila CFTR (Dmel\CFTR). We show that knockdown of Dmel\CFTR in the adult intestine disrupts osmotic homeostasis and displays CF-like phenotypes that lead to intestinal stem cell hyperplasia. We also show that expression of wild-type human CFTR, but not mutant variants of CFTR that prevent plasma membrane expression, rescues the mutant phenotypes of Dmel\CFTR Furthermore, we performed RNA sequencing (RNA-Seq)-based transcriptomic analysis using Dmel\CFTR fly intestine and identified a mucin gene, Muc68D, which is required for proper intestinal barrier protection. Altogether, our findings suggest that Drosophila can be a powerful model organism for studying CF pathophysiology.
Collapse
|
44
|
Davoodi S, Foley E. Host-Microbe-Pathogen Interactions: A Review of Vibrio cholerae Pathogenesis in Drosophila. Front Immunol 2020; 10:3128. [PMID: 32038640 PMCID: PMC6993214 DOI: 10.3389/fimmu.2019.03128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022] Open
Abstract
Most animals maintain mutually beneficial symbiotic relationships with their intestinal microbiota. Resident microbes in the gastrointestinal tract breakdown indigestible food, provide essential nutrients, and, act as a barrier against invading microbes, such as the enteric pathogen Vibrio cholerae. Over the last decades, our knowledge of V. cholerae pathogenesis, colonization, and transmission has increased tremendously. A number of animal models have been used to study how V. cholerae interacts with host-derived resources to support gastrointestinal colonization. Here, we review studies on host-microbe interactions and how infection with V. cholerae disrupts these interactions, with a focus on contributions from the Drosophila melanogaster model. We will discuss studies that highlight the connections between symbiont, host, and V. cholerae metabolism; crosstalk between V. cholerae and host microbes; and the impact of the host immune system on the lethality of V. cholerae infection. These studies suggest that V. cholerae modulates host immune-metabolic responses in the fly and improves Vibrio fitness through competition with intestinal microbes.
Collapse
Affiliation(s)
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
45
|
Özbek R, Wielsch N, Vogel H, Lochnit G, Foerster F, Vilcinskas A, von Reumont BM. Proteo-Transcriptomic Characterization of the Venom from the Endoparasitoid Wasp Pimpla turionellae with Aspects on Its Biology and Evolution. Toxins (Basel) 2019; 11:E721. [PMID: 31835557 PMCID: PMC6950128 DOI: 10.3390/toxins11120721] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/03/2019] [Accepted: 12/07/2019] [Indexed: 12/23/2022] Open
Abstract
Within mega-diverse Hymenoptera, non-aculeate parasitic wasps represent 75% of all hymenopteran species. Their ovipositor dual-functionally injects venom and employs eggs into (endoparasitoids) or onto (ectoparasitoids) diverse host species. Few endoparasitoid wasps such as Pimpla turionellae paralyze the host and suppress its immune responses, such as encapsulation and melanization, to guarantee their offspring's survival. Here, the venom and its possible biology and function of P. turionellae are characterized in comparison to the few existing proteo-transcriptomic analyses on parasitoid wasp venoms. Multiple transcriptome assembly and custom-tailored search and annotation strategies were applied to identify parasitoid venom proteins. To avoid false-positive hits, only transcripts were finally discussed that survived strict filter settings, including the presence in the proteome and higher expression in the venom gland. P. turionella features a venom that is mostly composed of known, typical parasitoid enzymes, cysteine-rich peptides, and other proteins and peptides. Several venom proteins were identified and named, such as pimplin2, 3, and 4. However, the specification of many novel candidates remains difficult, and annotations ambiguous. Interestingly, we do not find pimplin, a paralytic factor in Pimpla hypochondriaca, but instead a new cysteine inhibitor knot (ICK) family (pimplin2), which is highly similar to known, neurotoxic asilid1 sequences from robber flies.
Collapse
Affiliation(s)
- Rabia Özbek
- Project group Bioressources, Animal Venomics, Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchesterstrasse 2, 35392 Giessen, Germany
| | - Natalie Wielsch
- Research Group Mass Spectrometry/Proteomics, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745 Jena, Germany
| | - Heiko Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745 Jena, Germany;
| | - Günter Lochnit
- Protein Analytics, Institute of Biochemistry, Justus Liebig University, Friedrichstrasse 24, 35392 Giessen, Germany;
| | - Frank Foerster
- Bioinformatics Core Facility, Bioinformatics and Systems Biology, Justus Liebig University, Heinrich Buff Ring 58, 35394 Giessen, Germany
| | - Andreas Vilcinskas
- Project group Bioressources, Animal Venomics, Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchesterstrasse 2, 35392 Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich Buff Ring 58, 35394 Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| | - Björn Marcus von Reumont
- Project group Bioressources, Animal Venomics, Fraunhofer Institute for Molecular Biology and Applied Ecology, Winchesterstrasse 2, 35392 Giessen, Germany
- Institute for Insect Biotechnology, Justus Liebig University, Heinrich Buff Ring 58, 35394 Giessen, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| |
Collapse
|
46
|
Abstract
Regenerative processes that maintain the function of the gastrointestinal (GI) epithelium are critical for health and survival of multicellular organisms. In insects and vertebrates, intestinal stem cells (ISCs) regenerate the GI epithelium. ISC function is regulated by intrinsic, local, and systemic stimuli to adjust regeneration to tissue demands. These control mechanisms decline with age, resulting in significant perturbation of intestinal homeostasis. Processes that lead to this decline have been explored intensively in Drosophila melanogaster in recent years and are now starting to be characterized in mammalian models. This review presents a model for age-related regenerative decline in the fly intestine and discusses recent findings that start to establish molecular mechanisms of age-related decline of mammalian ISC function.
Collapse
Affiliation(s)
- Heinrich Jasper
- Immunology Discovery, Genentech, Inc., South San Francisco, California 94080, USA;
| |
Collapse
|
47
|
Liu A, Huang X, Gong L, Guo Z, Zhang Y, Yang Z. Characterization of immune-related PGRP gene expression and phenoloxidase activity in Cry1Ac-susceptible and -resistant Plutella xylostella (L.). PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 160:79-86. [PMID: 31519260 DOI: 10.1016/j.pestbp.2019.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 06/10/2023]
Abstract
Peptidoglycan recognition proteins (PGRPs) are important recognition receptors which play a critical role in signal identification and transmission in Toll or immune deficiency (IMD) pathways, particularly when pathogens evade and circumvent reactive oxygen species. Antimicrobial peptides (AMPs) synthesis can be activated by these signals to further eliminate pathogens. In this study, we cloned and characterized three different PGRP genes in Plutella xylostella strains, DBM1Ac-S, DBM1Ac-R and a field strain (DBMF). The results showed that PGRP1 belongs to the PGRP-SA family, PGRP2 to PGRP-LB, and PGRP3 to PGRP-LF. Moreover, PGRP1 expressed the highest transcript level, followed by PGRP3 and PGRP2, in two tissues including the gut and the larval carcass tissues of the DBM1Ac-S strain. Furthermore, altered expression levels of PGRP1-3 genes were detected in both gut and carcass tissues. Moreover, the DBM1Ac-R strain had the highest phenol oxidase (PO) activity among these three strains. The characterization of PGRP gene expression and PO activity in DBM1Ac-S, DBM1Ac-R and DBM-F provides insights into their important physiological roles in the immune system of P. xylostella exposed to Bt Cry1Ac toxin.
Collapse
Affiliation(s)
- Ao Liu
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, College of Plant Protection, Hunan Agricultural University, Changsha, China
| | - Xiufang Huang
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, College of Plant Protection, Hunan Agricultural University, Changsha, China
| | - Lijun Gong
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhaojiang Guo
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Youjun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhongxia Yang
- Hunan Provincial Key Laboratory for Biology and Control of Plant Diseases and Insect Pests, College of Plant Protection, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
48
|
Participation of the Serine Protease Jonah66Ci in the Drosophila Antinematode Immune Response. Infect Immun 2019; 87:IAI.00094-19. [PMID: 31182620 DOI: 10.1128/iai.00094-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/29/2019] [Indexed: 01/27/2023] Open
Abstract
Serine proteases and serine protease homologs form the second largest gene family in the Drosophila melanogaster genome. Certain genes in the Jonah multigene family encoding serine proteases have been implicated in the fly antiviral immune response. Here, we report the involvement of Jonah66Ci in the Drosophila immune defense against Steinernema carpocapsae nematode infection. We find that Drosophila Jonah66Ci is upregulated in response to symbiotic (carrying the mutualistic bacterium Xenorhabdus nematophila) or axenic (lacking Xenorhabdus) Steinernema nematodes and is expressed exclusively in the gut of Drosophila larvae. Inactivation of Jonah66Ci provides a survival advantage to larvae against axenic nematodes and results in differential expression of Toll and Imd pathway effector genes, specifically in the gut. Also, inactivation of Jonah66Ci increases the numbers of enteroendocrine and mitotic cells in the gut of uninfected larvae, and infection with Steinernema nematodes reduces their numbers, whereas the numbers of intestinal stem cells are unaffected by nematode infection. Jonah66Ci knockdown further reduces nitric oxide levels in response to infection with symbiotic Steinernema nematodes. Finally, we show that Jonah66Ci knockdown does not alter the feeding rates of uninfected Drosophila larvae; however, infection with axenic Steinernema nematodes lowers larval feeding. In conclusion, we report that Jonah66Ci participates in maintaining homeostasis of certain physiological processes in Drosophila larvae in the context of Steinernema nematode infection. Similar findings will take us a step further toward understanding the molecular and physiological mechanisms that take place during parasitic nematode infection in insects.
Collapse
|
49
|
Erlandson MA, Toprak U, Hegedus DD. Role of the peritrophic matrix in insect-pathogen interactions. JOURNAL OF INSECT PHYSIOLOGY 2019; 117:103894. [PMID: 31175854 DOI: 10.1016/j.jinsphys.2019.103894] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/27/2019] [Accepted: 06/05/2019] [Indexed: 05/12/2023]
Abstract
The peritrophic matrix (PM) is an acellular chitin and glycoprotein layer that lines the invertebrate midgut. The PM has long been considered a physical as well as a biochemical barrier, protecting the midgut epithelium from abrasive food particles, digestive enzymes and pathogens infectious per os. This short review will focus on the latter function, as a barrier to pathogens infectious per os. We focus on the evidence confirming the role of the PM as protective barrier against pathogenic microorganisms of insects, mainly bacteria and viruses, as well as the evolution of a variety of mechanisms used by pathogens to overcome the PM barrier.
Collapse
Affiliation(s)
- Martin A Erlandson
- Saskatoon Research and Development Centre, Agriculture and Agri-Food Canada, Saskatoon, Saskatchewan, Canada; Department of Biology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Umut Toprak
- Molecular Entomology Laboratory, Faculty of Agriculture, Ankara University, Ankara, Turkey
| | - Dwayne D Hegedus
- Saskatoon Research and Development Centre, Agriculture and Agri-Food Canada, Saskatoon, Saskatchewan, Canada; Department of Food and Bioproduct Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
50
|
Mergaert P. Role of antimicrobial peptides in controlling symbiotic bacterial populations. Nat Prod Rep 2019; 35:336-356. [PMID: 29393944 DOI: 10.1039/c7np00056a] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Covering: up to 2018 Antimicrobial peptides (AMPs) have been known for well over three decades as crucial mediators of the innate immune response in animals and plants, where they are involved in the killing of infecting microbes. However, AMPs have now also been found to be produced by eukaryotic hosts during symbiotic interactions with bacteria. These symbiotic AMPs target the symbionts and therefore have a more subtle biological role: not eliminating the microbial symbiont population but rather keeping it in check. The arsenal of AMPs and the symbionts' adaptations to resist them are in a careful balance, which contributes to the establishment of the host-microbe homeostasis. Although in many cases the biological roles of symbiotic AMPs remain elusive, for a number of symbiotic interactions, precise functions have been assigned or proposed to the AMPs, which are discussed here. The microbiota living on epithelia in animals, from the most primitive ones to the mammals, are challenged by a cocktail of AMPs that determine the specific composition of the bacterial community as well as its spatial organization. In the symbiosis of legume plants with nitrogen-fixing rhizobium bacteria, the host deploys an extremely large panel of AMPs - called nodule-specific cysteine-rich (NCR) peptides - that drive the bacteria into a terminally differentiated state and manipulate the symbiont physiology to maximize the benefit for the host. The NCR peptides are used as tools to enslave the bacterial symbionts, limiting their reproduction but keeping them metabolically active for nitrogen fixation. In the nutritional symbiotic interactions of insects and protists that have vertically transmitted bacterial symbionts with reduced genomes, symbiotic AMPs could facilitate the integration of the endosymbiont and host metabolism by favouring the flow of metabolites across the symbiont membrane through membrane permeabilization.
Collapse
Affiliation(s)
- P Mergaert
- Institute for Integrative Biology of the Cell, UMR9198, CNRS, Université Paris-Sud, CEA, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France.
| |
Collapse
|