1
|
Samantsidis GR, Karim S, Smith RC. Influence of blood feeding and infection on arthropod hemocytes. CURRENT OPINION IN INSECT SCIENCE 2025; 69:101341. [PMID: 39938680 DOI: 10.1016/j.cois.2025.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/14/2025]
Abstract
Blood feeding provides essential nutrients for development and reproduction in hematophagous arthropods yet also initiates significant other physiological alterations in immune function. Immune cells, or hemocytes, are integral components of the arthropod innate immune system with notable roles in defining vector competence. Evidence suggests that both blood feeding and infection drive substantial changes in hemocyte phenotypes, including proliferation, immune activation, and differentiation, which directly and indirectly influence pathogen infection outcomes. These dynamics have fueled extensive research into hemocyte biology in recent years, which aided by emerging single-cell technologies and methods of phagocyte depletion, have provided novel molecular insights into hemocyte populations and additional support for their important contributions to parasite, virus, and bacterial infections. Despite this progress, many aspects of arthropod immune cell biology remain unclear. Focusing on mosquitoes and ticks as two of the most prominent and well-studied arthropod vectors, this review summarizes the effects of blood feeding and infection on mosquito and tick hemocytes, highlighting hemocyte classifications, and the known mechanisms by which hemocytes can have positive or negative impacts on vector-borne pathogen infection.
Collapse
Affiliation(s)
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| | - Ryan C Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA.
| |
Collapse
|
2
|
Yu T, Li Y, Wang W, Lian X, Cheng J, Zuo J, Wang L, Song L. Treatment with inactivated Metschnikowia bicuspidata protects against the later infection in Chinese mitten crab Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2025:110421. [PMID: 40389102 DOI: 10.1016/j.fsi.2025.110421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
In recent years, infection by the fungus Metschnikowia bicuspidata has caused significant mortality in Chinese mitten crab Eriocheir sinensis and substantial economic losses, leaving effective control measures still unavailable. The protective effect of immune priming against the secondary bacterial infection reported in crabs provides a promising idea for the development of disease prevention strategies against M. bicuspidate invasion. This study demonstrated that the survival rate and immune capability of haemocytes in crabs pre-treated with inactivated M. bicuspidata were considerably enhanced following exposure to living M. bicuspidata. At 12 h after live M. bicuspidata challenge, the total haemocyte counts and proportion of new-born (EdU-positive) haemocytes all significantly increased compared with un-challenged group (p < 0.05), with markedly elevated levels in the M. bicuspidata pre-treated group relative to the untreated group (p < 0.05). The phagocytic rates of haemocytes to M. bicuspidata or latex bead also significantly increased in the pre-treated group (p < 0.05), accompanied by a higher level of reactive oxygen species than that in un-treated group. Specifically, among the three subtypes of haemocytes (granulocyte, semi-granulocyte, and agranulocyte), the percentage of agranulocytes (53.77%) was significantly (p < 0.05) higher in pre-treated group than that of the untreated group (42.12%), coinciding with the highest phagocytic rate in agranulocytes. The activities of phenoloxidase (PO) and lysozyme in the haemolymph supernatant significantly increased in the pre-treated group (p < 0.05), which also exhibited stronger inhibitory activity against the growth of M. bicuspidate compared to the untreated group. The expression levels of immune-related genes (Toll1 and ALF2) in the pre-treated group significantly increased (p < 0.05) following the live M. bicuspidata exposure. The results collectively indicated that treatment with inactivated M. bicuspidata can induce immune priming protection in E. sinensis, offering a novel and safe method to protect E. sinensis against infection of M. bicuspidata.
Collapse
Affiliation(s)
- Tianqi Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Yali Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China.
| | - Xingye Lian
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Junlei Cheng
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Jiajun Zuo
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
3
|
Saab SA, Cardoso-Jaime V, Kefi M, Dimopoulos G. Advances in the dissection of Anopheles-Plasmodium interactions. PLoS Pathog 2025; 21:e1012965. [PMID: 40163471 PMCID: PMC11957333 DOI: 10.1371/journal.ppat.1012965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Malaria is a life-threatening mosquito-borne disease caused by the Plasmodium parasite, responsible for more than half a million deaths annually and principally involving children. The successful transmission of malaria by Anopheles mosquitoes relies on complex successive interactions between the parasite and various mosquito organs, host factors, and restriction factors. This review summarizes our current understanding of the mechanisms regulating Plasmodium infection of the mosquito vector at successive plasmodial developmental stages and highlights potential transmission-blocking targets and strategies.
Collapse
Affiliation(s)
- Sally A. Saab
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Mary Kefi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| |
Collapse
|
4
|
Haraji S, Shahmohammadi N, Talaei-Hassanloui R, Jin G, Ahsan SM, Kim HJ, Choi HW, Jeon Y, Kwon M, Lee D, Kim Y. Lipoxin B 4 and lipocalin play a crucial role in insect immune-priming induced by a gut microbial commensal. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 163:105317. [PMID: 39798858 DOI: 10.1016/j.dci.2025.105317] [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: 11/19/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Host cabbage possesses an endophyte, Bacillus subtilis, which induced immune-priming of the diamondback moth, Plutella xylostella. In contrast, larvae raised under axenic conditions lost the chance to feed the bacteria and were highly susceptible to various pathogens. Addition of B. subtilis to axenic larvae significantly restored immune responses and enhanced survival rates following pathogen infections. The immune-priming factor(s) was determined among 18 apolipoprotein D (ApoD) genes identified as lipocalin candidates in P. xylostella, in which ApoD1 expression was functionally linked with B. subtilis-induced immune-priming. In addition, lipoxins were analyzed in immune-primed larvae via LC-MS/MS, in which LXB4 was detected, but not LXA4. The LXB4 titer was significantly higher than that that in the larvae reared under axenic conditions. Notably, LXB4 alone sufficiently induced significant immune responses. To support lipoxin biosynthesis in insects, this study identified a lipoxygenase-like peroxidase gene, HemP2. Its expression was induced in the immune-primed larvae. However, its suppression prevented LXB4 production under the immune-priming conditions. To explain the up-regulations of lipocalin/lipoxin by the gut commensal, Toll and IMD immune signaling pathways were analyzed. The up-regulation of ApoD1 and HemP2 expressions was mediated through the IMD, but not the Toll, immune signaling pathway in the larval gut of P. xylostella under B. subtilis-induced immune-priming conditions. This study highlights the potential role of commensal gut microbes including B. subtilis in driving immune-priming via an insect lipoxin-lipocalin complex through the IMD immune signaling pathway.
Collapse
Affiliation(s)
- Shiva Haraji
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea; Department of Plant Protection, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Niayesh Shahmohammadi
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Reza Talaei-Hassanloui
- Department of Plant Protection, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | - Gahyeon Jin
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - S M Ahsan
- Department of Plant Protection, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Hee-Jin Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Hyong Woo Choi
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Yongho Jeon
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Minji Kwon
- Industry Academy Cooperation Foundation, Andong National University, Andong, 36729, South Korea
| | - Donghee Lee
- Industry Academy Cooperation Foundation, Andong National University, Andong, 36729, South Korea
| | - Yonggyun Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea.
| |
Collapse
|
5
|
Rajendran D, Vinayagam S, Sekar K, Bhowmick IP, Sattu K. Symbiotic Bacteria: Wolbachia, Midgut Microbiota in Mosquitoes and Their Importance for Vector Prevention Strategies. MICROBIAL ECOLOGY 2024; 87:154. [PMID: 39681734 PMCID: PMC11649735 DOI: 10.1007/s00248-024-02444-6] [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: 12/22/2023] [Accepted: 04/02/2024] [Indexed: 12/18/2024]
Abstract
Mosquito-borne illnesses pose a significant threat to eradication under existing vector management measures. Chemo-based vector control strategies (use of insecticides) raise a complication of resistance and environmental pollution. Biological control methods are an alternative approach to overcoming this complication arising from insecticides. The mosquito gut microbiome is essential to supporting the factors that involve metabolic regulation and metamorphic development (from juvenile to adult), as well as the induction of an immune response. The induced immune response includes the JAK-STAT, IMD, and Toll pathways due to the microbial interaction with the midgut cells (MG cells) that prevent disease transmission to humans. The aforementioned sequel to the review provides information about endosymbiont Wolbachia, which contaminates insect cells, including germline and somatic cytoplasm, and inhibits disease-causing pathogen development and transmission by competing for resources within the cell. Moreover, it reduces the host population via cytoplasmic incompatibility (CI), feminization, male killing, and parthenogenesis. Furthermore, the Cif factor in Wolbachia is responsible for CI induction that produces inviable cells with the translocating systems and the embryonic defect-causing protein factor, WalE1 (WD0830), which manipulates the host actin. This potential of Wolbachia can be used to design a paratransgenic system to control vectors in the field. An extracellular symbiotic bacterium such as Asaia, which is grown in the growth medium, is used to transfer lethal genes within itself. Besides, the genetically transferred symbiotic bacteria infect the wild mosquito population and are easily manifold. So, it might be suitable for vector control strategies in the future.
Collapse
Affiliation(s)
- Devianjana Rajendran
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Sathishkumar Vinayagam
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Kathirvel Sekar
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Ipsita Pal Bhowmick
- Department of Malariology, ICMR-RMRCNE Region, Dibrugarh, Assam, 786010, India
| | - Kamaraj Sattu
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India.
| |
Collapse
|
6
|
Haraji S, Talaei-Hassanloui R, Ahmed S, Jin G, Lee D, Kim Y. Apolipoprotein D3 and LOX product play a role in immune-priming of a lepidopteran insect, Spodoptera exigua. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 158:105198. [PMID: 38795942 DOI: 10.1016/j.dci.2024.105198] [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: 04/19/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024]
Abstract
Immune-priming occurs in insects after a prior pathogen exposure. However, its underlying mechanism in insects remains elusive. In the present work, immune-priming was detected in a lepidopteran insect, Spodoptera exigua. Specifically, a prior infection with a heat-killed pathogenic bacterium, Escherichia coli, led to increased survival upon the second infection of different pathogens. Plasma collected from larvae with the prior infection possessed the immune-priming factor(s) that significantly up-regulated cellular and humoral immune responses of naïve larvae. Our study also finds that variations in the timing of plasma collection for priming larvae resulted in distinct impacts on both cellular and humoral responses. However, when the active plasma exhibiting the immune-priming was heat-treated, it lost this priming activity, therefore suggesting that protein factor(s) play a role in this immune-priming. An immunofluorescence assay showed that the hemocytes collected from the immune-primed larvae highly reacted to a polyclonal antibody specific to a vertebrate lipocalin, apolipoprotein D (ApoD). Among 27 ApoD genes (Se-ApoD1 ∼ Se-ApoD27) of S. exigua, Se-ApoD3 was found to be highly induced during the immune-priming, in which it was shown to be expressed in hemocytes and fat body from a fluorescence in situ hybridization analysis. RNA interference of Se-ApoD3 expression significantly impaired the immune-priming of S. exigua larvae. Moreover, the inhibition of eicosanoid biosynthesis suppressed the immune-priming, in which treatment with a lipoxygenase (LOX) inhibitor-and not treatment with a cyclooxygenase inhibitor-suppressed immune-priming. Further, an addition of LOX product such as lipoxin A4 or lipoxin B4 significantly rescued the lost immune-priming activity. Taken together, these results suggest that a complex of ApoD3 and LOX product mediates the immune-priming activity of S. exigua.
Collapse
Affiliation(s)
- Shiva Haraji
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea; Department of Plant Protection, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Reza Talaei-Hassanloui
- Department of Plant Protection, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran.
| | - Shabbir Ahmed
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Gahyeon Jin
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Donghee Lee
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea
| | - Yonggyun Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong, 36729, South Korea.
| |
Collapse
|
7
|
Kumar T, Maitra S, Rai R, Priyanka, Maitra S, Tirkey NN, Kumari R. The dichotomy between probiotic lactic acid bacteria and Plasmodium: A promising therapeutic avenue. Acta Trop 2024; 257:107284. [PMID: 38857820 DOI: 10.1016/j.actatropica.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Our understanding of gut microbial populations and their immense influence on host immunity, health, and diseases has increased deeply in recent years. Numerous reports have identified the role of mosquito and mammalian gut microbiota in the modulation of host susceptibility to Plasmodium infection. Artemisinin resistance in malaria-endemic regions necessitates the development of new, safer, and more affordable treatments to supplement existing therapies. In this review, we compiled a colossal amount of data from numerous studies that have assessed the roles played by gut microbial communities in Plasmodium infection, progression, transmission, and severity. Most interestingly, our study points to the overwhelming evidence from experimental studies in mural malaria to human trials, suggesting that the presence of lactic acid bacteria in the gut microbiota of mammalian hosts provides a great degree of protection against malaria. Therefore, our study provides a compelling narrative for probiotic administration as an adjunct therapy for combatting malaria.
Collapse
Affiliation(s)
- Tarkeshwar Kumar
- Department of Zoology, Panch Pargana Kisan College, Ranchi University, Ranchi, Jharkhand, 835204, India.
| | - Satarupa Maitra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Richa Rai
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Priyanka
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Satwat Maitra
- Noida International Institute of Medical Sciences, Greater Noida, Uttar Pradesh, India
| | | | - Rajesh Kumari
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
8
|
Prakash A, Fenner F, Shit B, Salminen TS, Monteith KM, Khan I, Vale PF. IMD-mediated innate immune priming increases Drosophila survival and reduces pathogen transmission. PLoS Pathog 2024; 20:e1012308. [PMID: 38857285 PMCID: PMC11192365 DOI: 10.1371/journal.ppat.1012308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/21/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
Invertebrates lack the immune machinery underlying vertebrate-like acquired immunity. However, in many insects past infection by the same pathogen can 'prime' the immune response, resulting in improved survival upon reinfection. Here, we investigated the mechanistic basis and epidemiological consequences of innate immune priming in the fruit fly Drosophila melanogaster when infected with the gram-negative bacterial pathogen Providencia rettgeri. We find that priming in response to P. rettgeri infection is a long-lasting and sexually dimorphic response. We further explore the epidemiological consequences of immune priming and find it has the potential to curtail pathogen transmission by reducing pathogen shedding and spread. The enhanced survival of individuals previously exposed to a non-lethal bacterial inoculum coincided with a transient decrease in bacterial loads, and we provide strong evidence that the effect of priming requires the IMD-responsive antimicrobial-peptide Diptericin-B in the fat body. Further, we show that while Diptericin B is the main effector of bacterial clearance, it is not sufficient for immune priming, which requires regulation of IMD by peptidoglycan recognition proteins. This work underscores the plasticity and complexity of invertebrate responses to infection, providing novel experimental evidence for the effects of innate immune priming on population-level epidemiological outcomes.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Florence Fenner
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Tiina S. Salminen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Katy M. Monteith
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Pedro F. Vale
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Patel NF, Oliver SV. Generation of specific immune memory by bacterial exposure in the major malaria vector Anopheles arabiensis (Diptera: Culicidae). CURRENT RESEARCH IN INSECT SCIENCE 2024; 5:100085. [PMID: 38779142 PMCID: PMC11109336 DOI: 10.1016/j.cris.2024.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
There is a growing body of evidence that invertebrates can generate improved secondary responses after a primary challenge. This immunological memory can be primed by a range of pathogens, including bacteria. The generation of immunological memory has been demonstrated in mosquitoes, with the memory primed by a range of initial stimuli. This study aimed to examine whether insecticide resistance affects the capacity to generate immunological memory. The primary hypothesis was tested by examining the capacity of genetically related laboratory-reared Anopheles arabiensis strains that differ by insecticide resistant phenotype to generate immunological memory. The competing hypothesis tested was that the bacterial virulence was the key determinant in generating immunological memory. Immune memory was generated in F1 females but not males. Immunological memory was demonstrated in both laboratory strains, but the efficacy differed by the insecticide resistant phenotype of the strain. An initial oral challenge provided by a blood meal resulted generated better memory than an oral challenge by sugar. The efficacy of memory generation between the two bacterial strains differed between the two mosquito strains. Regardless of the challenge, the two strains differed in their capacity to generate memory. This study therefore demonstrated that insecticide resistant phenotype affected the capacity of the two strains to generate immunological memory. Although this study needs to be replicated with wild mosquitoes, it does suggest that a potential role for insecticide resistance in the functioning of the immune system and memory generation of An. arabiensis.
Collapse
Affiliation(s)
- Nashrin F Patel
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Shüné V Oliver
- Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
10
|
Samantsidis GR, Kwon H, Wendland M, Fonder C, Smith RC. TNF signaling mediates cellular immune function and promotes malaria parasite killing in the mosquito Anopheles gambiae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592209. [PMID: 38746363 PMCID: PMC11092648 DOI: 10.1101/2024.05.02.592209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Tumor Necrosis Factor-α (TNF-α) is a proinflammatory cytokine and a master regulator of immune cell function in vertebrates. While previous studies have implicated TNF signaling in invertebrate immunity, the roles of TNF in mosquito innate immunity and vector competence have yet to be explored. Herein, we confirm the identification of a conserved TNF-α pathway in Anopheles gambiae consisting of the TNF-α ligand, Eiger, and its cognate receptors Wengen and Grindelwald. Through gene expression analysis, RNAi, and in vivo injection of recombinant TNF-α, we provide direct evidence for the requirement of TNF signaling in regulating mosquito immune cell function by promoting granulocyte midgut attachment, increased granulocyte abundance, and oenocytoid rupture. Moreover, our data demonstrate that TNF signaling is an integral component of anti-Plasmodium immunity that limits malaria parasite survival. Together, our data support the existence of a highly conserved TNF signaling pathway in mosquitoes that mediates cellular immunity and influences Plasmodium infection outcomes, offering potential new approaches to interfere with malaria transmission by targeting the mosquito host.
Collapse
Affiliation(s)
| | - Hyeogsun Kwon
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| | - Megan Wendland
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| | - Catherine Fonder
- Molecular, Cellular and Developmental Biology Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| |
Collapse
|
11
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
12
|
Silva RCMC, Gomes FM. Evolution of the Major Components of Innate Immunity in Animals. J Mol Evol 2024; 92:3-20. [PMID: 38281163 DOI: 10.1007/s00239-024-10155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 01/30/2024]
Abstract
Innate immunity is present in all animals. In this review, we explore the main conserved mechanisms of recognition and innate immune responses among animals. In this sense, we discuss the receptors, critical for binding to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs); the downstream signaling proteins; and transcription factors that govern immune responses. We also highlight conserved inflammatory mediators that are induced after the recognition of DAMPs and PAMPs. At last, we discuss the mechanisms that are involved in the regulation and/or generation of reactive oxygen species (ROS), influencing immune responses, like heme-oxygenases (HOs).
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Fábio Mendonça Gomes
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Adegoke A, Ribeiro JMC, Smith R, Karim S. Tick innate immune responses to hematophagy and Ehrlichia infection at single-cell resolution. Front Immunol 2024; 14:1305976. [PMID: 38274813 PMCID: PMC10808623 DOI: 10.3389/fimmu.2023.1305976] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Ticks rely on robust cellular and humoral responses to control microbial infection. However, several aspects of the tick's innate immune system remain uncharacterized, most notably that of the immune cells (called hemocytes), which are known to play a significant role in cellular and humoral responses. Despite the importance of hemocytes in regulating microbial infection, our understanding of their basic biology and molecular mechanisms remains limited. Therefore, we believe that a more detailed understanding of the role of hemocytes in the interactions between ticks and tick-borne microbes is crucial to illuminating their function in vector competence and to help identify novel targets for developing new strategies to block tick-borne pathogen transmission. Methods This study examined hemocytes from the lone star tick (Amblyomma americanum) at the transcriptomic level using the 10X genomics single-cell RNA sequencing platform to analyze hemocyte populations from unfed, partially blood-fed, and Ehrlichia chaffeensis-infected ticks. The functional role of differentially expressed hemocyte markers in hemocyte proliferation and Ehrlichia dissemination was determined using an RNA interference approach. Results and discussion Our data exhibit the identification of fourteen distinct hemocyte populations. Our results uncover seven distinct lineages present in uninfected and Ehrlichia-infected hemocyte clusters. The functional characterization of hemocytin, cystatin, fibronectin, and lipocalin demonstrate their role in hemocyte population changes, proliferation, and Ehrlichia dissemination. Conclusion Our results uncover the tick immune responses to Ehrlichia infection and hematophagy at a single-cell resolution. This work opens a new field of tick innate immunobiology to understand the role of hemocytes, particularly in response to prolonged blood-feeding (hematophagy), and tick-microbial interactions.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
14
|
Cime-Castillo J, Vargas V, Hernández-Tablas JM, Quezada-Ruiz E, Díaz G, Lanz-Mendoza H. The costs of transgenerational immune priming for homologous and heterologous infections with different serotypes of dengue virus in Aedes aegypti mosquitoes. Front Immunol 2023; 14:1286831. [PMID: 38170025 PMCID: PMC10760805 DOI: 10.3389/fimmu.2023.1286831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The immune system is a network of molecules, signaling pathways, transcription, and effector modulation that controls, mitigates, or eradicates agents that may affect the integrity of the host. In mosquitoes, the innate immune system is highly efficient at combating foreign organisms but has the capacity to tolerate vector-borne diseases. These implications lead to replication, dissemination, and ultimately the transmission of pathogenic organisms when feeding on a host. In recent years, it has been discovered that the innate immune response of mosquitoes can trigger an enhanced immunity response to the stimulus of a previously encountered pathogen. This phenomenon, called immune priming, is characterized by a molecular response that prevents the replication of viruses, parasites, or bacteria in the body. It has been documented that immune priming can be stimulated through homologous organisms or molecules, although it has also been documented that closely related pathogens can generate an enhanced immune response to a second stimulus with a related organism. However, the cost involved in this immune response has not been characterized through the transmission of the immunological experience from parents to offspring by transgenerational immune priming (TGIP) in mosquitoes. Here, we address the impact on the rates of oviposition, hatching, development, and immune response in Aedes aegypti mosquitoes, the mothers of which were stimulated with dengue virus serotypes 2 and/or 4, having found a cost of TGIP on the development time of the progeny of mothers with heterologous infections, with respect to mothers with homologous infections. Our results showed a significant effect on the sex ratio, with females being more abundant than males. We found a decrease in transcripts of the siRNA pathway in daughters of mothers who had been exposed to an immune challenge with DV. Our research demonstrates that there are costs and benefits associated with TGIP in Aedes aegypti mosquitoes exposed to DV. Specifically, priming results in a lower viral load in the offspring of mothers who have previously been infected with the virus. Although some results from tests of two dengue virus serotypes show similarities, such as the percentage of pupae emergence, there are differences in the percentage of adult emergence, indicating differences in TGIP costs even within the same virus with different serotypes. This finding has crucial implications in the context of dengue virus transmission in endemic areas where multiple serotypes circulate simultaneously.
Collapse
Affiliation(s)
- Jorge Cime-Castillo
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Valeria Vargas
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
- Biomedical Research Institute, Universidad Nacional Autonoma de México, Ciudad de México, Mexico
| | - Juan Manuel Hernández-Tablas
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Edgar Quezada-Ruiz
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Grecia Díaz
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| | - Humberto Lanz-Mendoza
- Infection and Immunity Direction/Vector Borne Disease Department, Centro de Investigaciones Sobre Enfermedades Infecciosas-Instituto Nacional de Salud Pública (INSP), Cuernavaca, Mexico
| |
Collapse
|
15
|
Wang Z, Yong H, Zhang S, Liu Z, Zhao Y. Colonization Resistance of Symbionts in Their Insect Hosts. INSECTS 2023; 14:594. [PMID: 37504600 PMCID: PMC10380809 DOI: 10.3390/insects14070594] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023]
Abstract
The symbiotic microbiome is critical in promoting insect resistance against colonization by exogenous microorganisms. The mechanisms by which symbionts contribute to the host's immune capacity is referred to as colonization resistance. Symbionts can protect insects from exogenous pathogens through a variety of mechanisms, including upregulating the expression of host immune-related genes, producing antimicrobial substances, and competitively excluding pathogens. Concordantly, insects have evolved fine-tuned regulatory mechanisms to avoid overactive immune responses against symbionts or specialized cells to harbor symbionts. Alternatively, some symbionts have evolved special adaptations, such as the formation of biofilms to increase their tolerance to host immune responses. Here, we provide a review of the mechanisms about colonization resistance of symbionts in their insect hosts. Adaptations of symbionts and their insect hosts that may maintain such symbiotic relationships, and the significance of such relationships in the coevolution of symbiotic systems are also discussed to provide insights into the in-depth study of the contribution of symbionts to host physiology and behavior.
Collapse
Affiliation(s)
- Zhengyan Wang
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Hanzi Yong
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Shan Zhang
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Zhiyuan Liu
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China
| | - Yaru Zhao
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| |
Collapse
|
16
|
Ratnayake OC, Chotiwan N, Saavedra-Rodriguez K, Perera R. The buzz in the field: the interaction between viruses, mosquitoes, and metabolism. Front Cell Infect Microbiol 2023; 13:1128577. [PMID: 37360524 PMCID: PMC10289420 DOI: 10.3389/fcimb.2023.1128577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 06/28/2023] Open
Abstract
Among many medically important pathogens, arboviruses like dengue, Zika and chikungunya cause severe health and economic burdens especially in developing countries. These viruses are primarily vectored by mosquitoes. Having surmounted geographical barriers and threat of control strategies, these vectors continue to conquer many areas of the globe exposing more than half of the world's population to these viruses. Unfortunately, no medical interventions have been capable so far to produce successful vaccines or antivirals against many of these viruses. Thus, vector control remains the fundamental strategy to prevent disease transmission. The long-established understanding regarding the replication of these viruses is that they reshape both human and mosquito host cellular membranes upon infection for their replicative benefit. This leads to or is a result of significant alterations in lipid metabolism. Metabolism involves complex chemical reactions in the body that are essential for general physiological functions and survival of an organism. Finely tuned metabolic homeostases are maintained in healthy organisms. However, a simple stimulus like a viral infection can alter this homeostatic landscape driving considerable phenotypic change. Better comprehension of these mechanisms can serve as innovative control strategies against these vectors and viruses. Here, we review the metabolic basis of fundamental mosquito biology and virus-vector interactions. The cited work provides compelling evidence that targeting metabolism can be a paradigm shift and provide potent tools for vector control as well as tools to answer many unresolved questions and gaps in the field of arbovirology.
Collapse
Affiliation(s)
- Oshani C. Ratnayake
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Nunya Chotiwan
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Karla Saavedra-Rodriguez
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Rushika Perera
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
17
|
|
18
|
Malaria oocysts require circumsporozoite protein to evade mosquito immunity. Nat Commun 2022; 13:3208. [PMID: 35680915 PMCID: PMC9184642 DOI: 10.1038/s41467-022-30988-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 05/25/2022] [Indexed: 11/08/2022] Open
Abstract
Malaria parasites are less vulnerable to mosquito immune responses once ookinetes transform into oocysts, facilitating parasite development in the mosquito. However, the underlying mechanisms of oocyst resistance to mosquito defenses remain unclear. Here, we show that circumsporozoite protein (CSP) is required for rodent malaria oocysts to avoid mosquito defenses. Mosquito infection with CSPmut parasites (mutation in the CSP pexel I/II domains) induces nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 5 (NOX5)-mediated hemocyte nitration, thus activating Toll pathway and melanization of mature oocysts, upregulating hemocyte TEP1 expression, and causing defects in the release of sporozoites from oocysts. The pre-infection of mosquitoes with the CSPmut parasites reduces the burden of infection when re-challenged with CSPwt parasites by inducing hemocyte nitration. Thus, we demonstrate why oocysts are invisible to mosquito immunity and reveal an unknown role of CSP in the immune evasion of oocysts, indicating it as a potential target to block malaria transmission. Circumsporozoite protein (CSP), the major surface protein of Plasmodium sporozoites, is important for parasite targeting to mosquito salivary glands and the mammalian liver. Here, Zhu et al. show that CSP is required for rodent malaria oocysts to evade mosquito immunity by inducing hemocyte nitration and causing subsequent defects in sporozoite-release from oocysts.
Collapse
|
19
|
Zhang H, Tan AR, Li PJ, Lu SP, Jia QC, Huang SN, Bai J, Hou YM. A specific primed immune response in red palm weevil, Rhynchophorus ferrugineus, is mediated by hemocyte differentiation and phagocytosis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 131:104380. [PMID: 35245605 DOI: 10.1016/j.dci.2022.104380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Red palm weevil, Rhynchophorus ferrugineus, is an invasive and destructive pest that causes serious damages to palm trees. Like other invertebrates, red palm weevil relies solely on its innate immune response to fight invading microbes; by definition, innate immunity lacks adaptive characteristics. However, we show here that priming the red palm weevil larvae with heat-killed Bacillus thuringiensis specifically increased survival of the larvae during a secondary lethal infection with live bacteria, and B. thuringiensis primed larvae also showed a higher clearance efficiency for this bacterium, which indicated that the red palm weevil larvae possessed a strong immune priming response. The degree of enhanced immune protection was positively correlated with hemocyte proliferation and the level of phagocytic ability of hemocytes. Moreover, the red palm weevil larvae primed by B. thuringiensis induced the continuous synthesis of serotonin in the hemolymph, which in turn enhanced the phagocytic ability and pathogen clearance ability of the host, representing an important mechanism for the red palm weevil to achieve priming protection. Our findings reveal a specific immune priming of the red palm weevil larvae mediated by the continuous secretion of serotonin, and provide new insights into the mechanisms of invertebrates immune priming.
Collapse
Affiliation(s)
- He Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Department of Biology, Xinzhou Teachers University, Xinzhou, Shanxi, 034000, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - An-Ran Tan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Peng-Ju Li
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Sheng-Ping Lu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Qing-Chen Jia
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Shu-Ning Huang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - Juan Bai
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China
| | - You-Ming Hou
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350002, PR China.
| |
Collapse
|
20
|
Wu G, Liu J, Li M, Xiao Y, Yi Y. Prior infection of Galleria mellonella with sublethal dose of Bt elicits immune priming responses but incurs metabolic changes. JOURNAL OF INSECT PHYSIOLOGY 2022; 139:104401. [PMID: 35636486 DOI: 10.1016/j.jinsphys.2022.104401] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
Invertebrate immune priming has attracted wide attention of biologists in recent years because it challenges core notions about the disparate nature of acquired and innate immunity. However, the metabolic switch and energetic cost during eliciting immune priming are poorly investigated issues, which could widen and deepen our understanding of the physiological mechanism of immune priming. In this study, using sublethal dose of Bacillus thuringiensis (Bt) as an elicitor, we detected typical immune priming responses in Galleria mellonella. We found that the intensity of immune priming is positively correlated with the levels of antimicrobial peptides and phagocytosis ability of hemocytes. Subsequently, we employed LC-MS/MS-based untargeted metabolomics techniques to analyze the metabolic changes in the fat body of G. mellonella larvae during immune priming. The results showed that there were 74 and 56 significantly altered metabolites in positive and negative ion mode, respectively, after Bt priming. Most of the differential metabolites were enriched in the following metabolic pathways: amino acid biosynthesis, carbon metabolism, aminoacyl-tRNA biosynthesis and ABC transporters. The energetic cost of immune priming was depicted mainly in the slow growth of body mass and decreased levels of sucrose, lactose, D-ribulose 1,5-bisphosphate, Glycerate-3P and isocitric acid, which are enriched in carbon metabolism and involved in energy production. Meanwhile, correlation and interaction network analysis showed negative correlations between carbohydrates and metabolites involved in amino acid biosynthesis, suggesting that amino acids acted as the main energy source and helped the organisms synthesize immune effectors to participate in the immune priming response. Our results pave the way for uncovering the physiological mechanism of insect immune priming and discovering novel targets for Bt insecticide.
Collapse
Affiliation(s)
- Gongqing Wu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China; Guangdong Cosmetics Engineering & Technology Research Center, Zhongshan 528458, PR China
| | - Jiajie Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China
| | - Mei Li
- University of Electronic Science and Technology of China, Zhongshan Institute, Zhongshan 528402, PR China
| | - Yang Xiao
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Science, Guangzhou 510610, PR China
| | - Yunhong Yi
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, PR China.
| |
Collapse
|
21
|
Shaw WR, Marcenac P, Catteruccia F. Plasmodium development in Anopheles: a tale of shared resources. Trends Parasitol 2022; 38:124-135. [PMID: 34548252 PMCID: PMC8758519 DOI: 10.1016/j.pt.2021.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Interactions between the Anopheles mosquito vector and Plasmodium parasites shape how malaria is transmitted in endemic regions. The long association of these two organisms has led to evolutionary processes that minimize fitness costs of infection and benefit both players through shared nutrient resources, parasite immune suppression, and mosquito tolerance to infection. In this review we explore recent data describing how Plasmodium falciparum, the deadliest malaria parasite, associates with one of its most important natural mosquito hosts, Anopheles gambiae, and we discuss the implications of these findings for parasite transmission and vector control strategies currently in development.
Collapse
Affiliation(s)
- W Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Perrine Marcenac
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
22
|
Liu QX, Su ZP, Liu HH, Lu SP, Zhao Y, Ma B, Hou YM, Shi ZH. Current understanding and perspectives on the potential mechanisms of immune priming in beetles. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104305. [PMID: 34718077 DOI: 10.1016/j.dci.2021.104305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
Beetles are the most diverse group of insects in Insecta which can be found in almost every habitat and environment on Earth. The possessing of the rapid and effective immune defenses is one of the important factors for their success. It is generally recognized that beetles only rely on the non-specific innate immune defense, without immunological memory, to fight against pathogens. However, there was cumulative evidence for the innate immune memory in invertebrates, including beetles, over the last decades, implying that insect innate immunity is more complex and has more features than previously thought. In beetles, it has been well documented that the specific or nonspecific enhanced immunocompetence can persist throughout development within generations and can even be transferred to the descendents in the next generation. Although insect immune priming might be shaped by epigenetic modifications and transferring effectors, mRNA and microbial signals, the solid experimental evidence to support the causal relationship between any of them and immune priming is still scarce. The combined usage of 'omics' approaches and CRISPR/Cas9 in the appropriate insect models with well-known genetic background, Tribolium castaneum and Tenebrio molitor, will help us to decipher the molecular mechanisms by which immune priming occurs in beetles in depth.
Collapse
Affiliation(s)
- Qian-Xia Liu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhi-Ping Su
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Hui-Hui Liu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Sheng-Ping Lu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yue Zhao
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Bing Ma
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - You-Ming Hou
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhang-Hong Shi
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
23
|
Ali Mohammadie Kojour M, Baliarsingh S, Jang HA, Yun K, Park KB, Lee JE, Han YS, Patnaik BB, Jo YH. Current knowledge of immune priming in invertebrates, emphasizing studies on Tenebrio molitor. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 127:104284. [PMID: 34619174 DOI: 10.1016/j.dci.2021.104284] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/16/2021] [Accepted: 10/01/2021] [Indexed: 06/13/2023]
Abstract
Vertebrates rely on the most sophisticated adaptive immunity to defend themselves against various pathogens. This includes immunologic memory cells, which mount a stronger and more effective immune response against an antigen after its first encounter. Unlike vertebrates, invertebrates' defense completely depends on the innate immunity mechanisms including humoral and cell-mediated immunity. Furthermore, the invertebrate equivalent of the memory cells was discovered only recently. Since the discovery of transgenerational immune priming (TGIP) in crustaceans, numerous findings have proven the IP in invertebrate classes such as insects. TGIP can be induced through maternal priming pathways such as transcriptional regulation of antimicrobial peptides, and also paternal IP including the induction of proPO system activity. We appraise the diversity and specificity of IP agents to provide sustained immunologic memory in insects, particularly T. molitor in the review. An understanding of IP (more so TGIP) response in T. molitor will deepen our knowledge of invertebrate immunity, and boost the mass-rearing industry by reducing pathogen infection rates.
Collapse
Affiliation(s)
- Maryam Ali Mohammadie Kojour
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Snigdha Baliarsingh
- PG Department of Biosciences and Biotechnology, Fakir Mohan University, Balasore, Odisha, 756089, India
| | - Ho Am Jang
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Keunho Yun
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Ki Beom Park
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Jong Eun Lee
- Department of Biological Science and Biotechnology, Andong National University, Andong, 36729, South Korea
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Bharat Bhusan Patnaik
- PG Department of Biosciences and Biotechnology, Fakir Mohan University, Balasore, Odisha, 756089, India.
| | - Yong Hun Jo
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
24
|
Prakash A, Khan I. Why do insects evolve immune priming? A search for crossroads. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104246. [PMID: 34453994 DOI: 10.1016/j.dci.2021.104246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
Until recently, it was assumed that insects lack immune memory since they do not have vertebrate-like specialized memory cells. Therefore, their most well studied evolutionary response against pathogens was increased basal immunity. However, growing evidence suggests that many insects also exhibit a form of immune memory (immune priming), where prior exposure to a low dose of infection confers protection against subsequent infection by the same pathogen that acts both within and across generations. Most strikingly, they can rapidly evolve as a highly parallel and mutually exclusive strategy from basal immunity, under different selective conditions and with divergent evolutionary trade-offs. However, the relative importance of priming as an optimal immune strategy also has contradictions, primarily because supporting mechanisms are still unclear. In this review, we adopt a comparative approach to highlight several emerging evolutionary, ecological and mechanistic features of priming vs basal immune responses that warrant immediate attention for future research.
Collapse
Affiliation(s)
- Arun Prakash
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, EH9 3FL, United Kingdom.
| | - Imroze Khan
- Department of Biology, Ashoka University, Plot No. 2, Rajiv Gandhi Education City, P.O. Rai, Sonepat, Haryana, 131029, India.
| |
Collapse
|
25
|
Prakash A, Agashe D, Khan I. The costs and benefits of basal infection resistance vs immune priming responses in an insect. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104261. [PMID: 34536466 DOI: 10.1016/j.dci.2021.104261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/31/2021] [Accepted: 09/14/2021] [Indexed: 06/13/2023]
Abstract
In insects, basal pathogen resistance and immune priming can evolve as mutually exclusive strategies, with distinct infection outcomes. However, the evolutionary drivers of such diverse immune functions remain poorly understood. Here, we addressed this key issue by systematically analyzing the differential fitness costs and benefits of priming vs resistance evolution in Tribolium beetle populations infected with Bacillus thuringiensis. Surprisingly, resistant beetles had increased post-infection reproduction and a longer lifespan under both starving as well as fed conditions, with no other measurable costs. In contrast, priming reduced offspring early survival, development rate and reproduction. Priming did improve post-infection survival of offspring, but this added trans-generational benefit of immune priming might not compensate for its pervasive costs. Resistance was thus consistently more beneficial. Overall, our work demonstrates the evolutionary change in trans-generational priming response, and provides a detailed comparison of the complex fitness consequences of evolved priming vs resistance.
Collapse
Affiliation(s)
- Arun Prakash
- National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560065, India
| | - Deepa Agashe
- National Centre for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560065, India.
| | - Imroze Khan
- Ashoka University, Plot No. 2, Rajiv Gandhi Education City, P.O. Rai, Sonepat, Haryana, 131029, India.
| |
Collapse
|
26
|
Double peroxidase and histone acetyltransferase AgTip60 maintain innate immune memory in primed mosquitoes. Proc Natl Acad Sci U S A 2021; 118:2114242118. [PMID: 34711682 DOI: 10.1073/pnas.2114242118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Immune priming in Anopheles gambiae is mediated by the systemic release of a hemocyte differentiation factor (HDF), a complex of lipoxin A4 bound to Evokin, a lipid carrier. HDF increases the proportion of circulating granulocytes and enhances mosquito cellular immunity. Here, we show that Evokin is present in hemocytes and fat-body cells, and messenger RNA (mRNA) expression increases significantly after immune priming. The double peroxidase (DBLOX) enzyme, present in insects but not in vertebrates, is essential for HDF synthesis. DBLOX is highly expressed in oenocytes in the fat-body tissue, and these cells increase in number in primed mosquitoes. We provide direct evidence that the histone acetyltransferase AgTip60 (AGAP001539) is also essential for a sustained increase in oenocyte numbers, HDF synthesis, and immune priming. We propose that oenocytes may function as a population of cells that are reprogrammed, and orchestrate and maintain a broad, systemic, and long-lasting state of enhanced immune surveillance in primed mosquitoes.
Collapse
|
27
|
Kwon H, Hall DR, Smith RC. Prostaglandin E2 Signaling Mediates Oenocytoid Immune Cell Function and Lysis, Limiting Bacteria and Plasmodium Oocyst Survival in Anopheles gambiae. Front Immunol 2021; 12:680020. [PMID: 34484178 PMCID: PMC8415482 DOI: 10.3389/fimmu.2021.680020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid-derived signaling molecules known as eicosanoids have integral roles in mediating immune and inflammatory processes across metazoans. This includes the function of prostaglandins and their cognate G protein-coupled receptors (GPCRs) to employ their immunological actions. In insects, prostaglandins have been implicated in the regulation of both cellular and humoral immune responses, yet in arthropods of medical importance, studies have been limited. Here, we describe a prostaglandin E2 receptor (AgPGE2R) in the mosquito Anopheles gambiae and demonstrate that its expression is most abundant in oenocytoid immune cell populations. Through the administration of prostaglandin E2 (PGE2) and AgPGE2R-silencing, we demonstrate that prostaglandin E2 signaling regulates a subset of prophenoloxidases (PPOs) and antimicrobial peptides (AMPs) that are strongly expressed in populations of oenocytoids. We demonstrate that PGE2 signaling via the AgPGE2R significantly limits both bacterial replication and Plasmodium oocyst survival. Additional experiments establish that PGE2 treatment increases phenoloxidase (PO) activity through the increased expression of PPO1 and PPO3, genes essential to anti-Plasmodium immune responses that promote oocyst killing. We also provide evidence that the mechanisms of PGE2 signaling are concentration-dependent, where high concentrations of PGE2 promote oenocytoid lysis, negating the protective effects of lower concentrations of PGE2 on anti-Plasmodium immunity. Taken together, our results provide new insights into the role of PGE2 signaling on immune cell function and its contributions to mosquito innate immunity that promote pathogen killing.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, IA, United States
| | - David R Hall
- Department of Entomology, Iowa State University, Ames, IA, United States
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, IA, United States
| |
Collapse
|
28
|
Kwon H, Mohammed M, Franzén O, Ankarklev J, Smith RC. Single-cell analysis of mosquito hemocytes identifies signatures of immune cell subtypes and cell differentiation. eLife 2021; 10:66192. [PMID: 34318744 PMCID: PMC8376254 DOI: 10.7554/elife.66192] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Mosquito immune cells, known as hemocytes, are integral to cellular and humoral responses that limit pathogen survival and mediate immune priming. However, without reliable cell markers and genetic tools, studies of mosquito immune cells have been limited to morphological observations, leaving several aspects of their biology uncharacterized. Here, we use single-cell RNA sequencing (scRNA-seq) to characterize mosquito immune cells, demonstrating an increased complexity to previously defined prohemocyte, oenocytoid, and granulocyte subtypes. Through functional assays relying on phagocytosis, phagocyte depletion, and RNA-FISH experiments, we define markers to accurately distinguish immune cell subtypes and provide evidence for immune cell maturation and differentiation. In addition, gene-silencing experiments demonstrate the importance of lozenge in defining the mosquito oenocytoid cell fate. Together, our scRNA-seq analysis provides an important foundation for future studies of mosquito immune cell biology and a valuable resource for comparative invertebrate immunology.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, United States
| | - Mubasher Mohammed
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Oscar Franzén
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Microbial Single Cell Genomics facility, SciLifeLab, Biomedical Center (BMC) Uppsala University, Uppsala, Sweden
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, United States
| |
Collapse
|
29
|
Kaczmarek A, Wrońska AK, Boguś MI, Kazek M, Gliniewicz A, Mikulak E, Matławska M. The type of blood used to feed Aedes aegypti females affects their cuticular and internal free fatty acid (FFA) profiles. PLoS One 2021; 16:e0251100. [PMID: 33930098 PMCID: PMC8087090 DOI: 10.1371/journal.pone.0251100] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/19/2021] [Indexed: 11/18/2022] Open
Abstract
Aedes aegypti, the primary vector of various arthropod-borne viral (arboviral) diseases such as dengue and Zika, is a popular laboratory model in vector biology. However, its maintenance in laboratory conditions is difficult, mostly because the females require blood meals to complete oogenesis, which is often provided as sheep blood. The outermost layer of the mosquito cuticle is consists of lipids which protects against numerous entomopathogens, prevents desiccation and plays an essential role in signalling processes. The aim of this work was to determine how the replacement of human blood with sheep blood affects the cuticular and internal FFA profiles of mosquitoes reared in laboratory culture. The individual FFAs present in cuticular and internal extracts from mosquito were identified and quantified by GC-MS method. The normality of their distribution was checked using the Kolmogorov-Smirnov test and the Student's t-test was used to compare them. GC-MS analysis revealed similar numbers of internal and cuticular FFAs in the female mosquitoes fed sheep blood by membrane (MFSB) and naturally fed human blood (NFHB), however MFSB group demonstrated 3.1 times greater FFA concentrations in the cuticular fraction and 1.4 times the internal fraction than the NFHB group. In the MFSB group, FFA concentration was 1.6 times higher in the cuticular than the internal fraction, while for NFHB, FFA concentration was 1.3 times lower in the cuticular than the internal fraction. The concentration of C18:3 acid was 223 times higher in the internal fraction than the cuticle in the MHSB group but was absent in the NFHB group. MFSB mosquito demonstrate different FFA profiles to wild mosquitoes, which might influence their fertility and the results of vital processes studied under laboratory conditions. The membrane method of feeding mosquitoes is popular, but our research indicates significant differences in the FFA profiles of MFSB and NFHB. Such changes in FFA profile might influence female fertility, as well as other vital processes studied in laboratory conditions, such as the response to pesticides. Our work indicates that sheep blood has potential shortcomings as a substitute feed for human blood, as its use in laboratory studies may yield different results to those demonstrated by free-living mosquitoes.
Collapse
Affiliation(s)
- Agata Kaczmarek
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- * E-mail:
| | | | - Mieczysława Irena Boguś
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- BIOMIBO, Warsaw, Poland
| | - Michalina Kazek
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | | | - Ewa Mikulak
- National Institute of Public Health—National Institute of Hygiene, Warsaw, Poland
| | - Marta Matławska
- National Institute of Public Health—National Institute of Hygiene, Warsaw, Poland
| |
Collapse
|
30
|
Dekmak AS, Yang X, Zu Dohna H, Buchon N, Osta MA. The Route of Infection Influences the Contribution of Key Immunity Genes to Antibacterial Defense in Anopheles gambiae. J Innate Immun 2020; 13:107-126. [PMID: 33207342 DOI: 10.1159/000511401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Insect systemic immune responses to bacterial infections have been mainly studied using microinjections, whereby the microbe is directly injected into the hemocoel. While this methodology has been instrumental in defining immune signaling pathways and enzymatic cascades in the hemolymph, it remains unclear whether and to what extent the contribution of systemic immune defenses to host microbial resistance varies if bacteria invade the hemolymph after crossing the midgut epithelium subsequent to an oral infection. Here, we address this question using the pathogenic Serratia marcescens (Sm) DB11 strain to establish systemic infections of the malaria vector Anopheles gambiae, either by septic Sm injections or by midgut crossing after feeding on Sm. Using functional genetic studies by RNAi, we report that the two humoral immune factors, thioester-containing protein 1 and C-type lectin 4, which play key roles in defense against Gram-negative bacterial infections, are essential for defense against systemic Sm infections established through injection, but they become dispensable when Sm infects the hemolymph following oral infection. Similar results were observed for the mosquito Rel2 pathway. Surprisingly, blocking phagocytosis by cytochalasin D treatment did not affect mosquito susceptibility to Sm infections established through either route. Transcriptomic analysis of mosquito midguts and abdomens by RNA-seq revealed that the transcriptional response in these tissues is more pronounced in response to feeding on Sm. Functional classification of differentially expressed transcripts identified metabolic genes as the most represented class in response to both routes of infection, while immune genes were poorly regulated in both routes. We also report that Sm oral infections are associated with significant downregulation of several immune genes belonging to different families, specifically the clip-domain serine protease family. In sum, our findings reveal that the route of infection not only alters the contribution of key immunity genes to host antimicrobial defense but is also associated with different transcriptional responses in midguts and abdomens, possibly reflecting different adaptive strategies of the host.
Collapse
Affiliation(s)
- Amira San Dekmak
- Biology Department, American University of Beirut, Beirut, Lebanon
| | - Xiaowei Yang
- Entomology Department, Cornell Institute for Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | | | - Nicolas Buchon
- Entomology Department, Cornell Institute for Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | - Mike A Osta
- Biology Department, American University of Beirut, Beirut, Lebanon,
| |
Collapse
|
31
|
Raddi G, Barletta ABF, Efremova M, Ramirez JL, Cantera R, Teichmann SA, Barillas-Mury C, Billker O. Mosquito cellular immunity at single-cell resolution. Science 2020; 369:1128-1132. [PMID: 32855340 DOI: 10.1126/science.abc0322] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/30/2020] [Indexed: 11/02/2022]
Abstract
Hemocytes limit the capacity of mosquitoes to transmit human pathogens. Here we profile the transcriptomes of 8506 hemocytes of Anopheles gambiae and Aedes aegypti mosquito vectors. Our data reveal the functional diversity of hemocytes, with different subtypes of granulocytes expressing distinct and evolutionarily conserved subsets of effector genes. A previously unidentified cell type in An. gambiae, which we term "megacyte," is defined by a specific transmembrane protein marker (TM7318) and high expression of lipopolysaccharide-induced tumor necrosis factor-α transcription factor 3 (LL3). Knockdown experiments indicate that LL3 mediates hemocyte differentiation during immune priming. We identify and validate two main hemocyte lineages and find evidence of proliferating granulocyte populations. This atlas of medically relevant invertebrate immune cells at single-cell resolution identifies cellular events that underpin mosquito immunity to malaria infection.
Collapse
Affiliation(s)
- Gianmarco Raddi
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 2AZ, UK.,Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ana Beatriz F Barletta
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | | | - Jose Luis Ramirez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Rafael Cantera
- Zoology Department, Stockholm University, Stockholm S-10691, Sweden.,Departamento de Biología del Neurodesarrollo, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 2AZ, UK.,Institute and Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Oliver Billker
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 2AZ, UK. .,Molecular Infection Medicine Sweden, Molecular Biology Department, Umeå University, Umeå S-90187, Sweden
| |
Collapse
|
32
|
Powers JC, Turangan R, Joosse BA, Hillyer JF. Adult Mosquitoes Infected with Bacteria Early in Life Have Stronger Antimicrobial Responses and More Hemocytes after Reinfection Later in Life. INSECTS 2020; 11:insects11060331. [PMID: 32481519 PMCID: PMC7349202 DOI: 10.3390/insects11060331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
The immunological strategies employed by insects to overcome infection vary with the type of infection and may change with experience. We investigated how a bacterial infection in the hemocoel of the African malaria mosquito, Anopheles gambiae, prepares the immune system to face a subsequent bacterial infection. For this, adult female mosquitoes were separated into three groups—unmanipulated, injured, or infected with Escherichia coli—and five days later all the mosquitoes were infected with a different strain of E. coli. We found that an injury or a bacterial infection early in life enhances the ability of mosquitoes to kill bacteria later in life. This protection results in higher mosquito survival and is associated with an increased hemocyte density, altered phagocytic activity by individual hemocytes, and the increased expression of nitric oxide synthase and perhaps prophenoloxidase 6. Protection from a second infection likely occurs because of heightened immune awareness due to an already existing infection instead of memory arising from an earlier, cured infection. This study highlights the dynamic nature of the mosquito immune response and how one infection prepares mosquitoes to survive a subsequent infection.
Collapse
|
33
|
Pondeville E, Puchot N, Parvy JP, Carissimo G, Poidevin M, Waterhouse RM, Marois E, Bourgouin C. Hemocyte-targeted gene expression in the female malaria mosquito using the hemolectin promoter from Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 120:103339. [PMID: 32105779 PMCID: PMC7181189 DOI: 10.1016/j.ibmb.2020.103339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Hemocytes, the immune cells in mosquitoes, participate in immune defenses against pathogens including malaria parasites. Mosquito hemocytes can also be infected by arthropod-borne viruses but the pro- or anti-viral nature of this interaction is unknown. Although there has been progress on hemocyte characterization during pathogen infection in mosquitoes, the specific contribution of hemocytes to immune responses and the hemocyte-specific functions of immune genes and pathways remain unresolved due to the lack of genetic tools to manipulate gene expression in these cells specifically. Here, we used the Gal4-UAS system to characterize the activity of the Drosophila hemocyte-specific hemolectin promoter in the adults of Anopheles gambiae, the malaria mosquito. We established an hml-Gal4 driver line that we further crossed to a fluorescent UAS responder line, and examined the expression pattern in the adult progeny driven by the hml promoter. We show that the hml regulatory region drives hemocyte-specific transgene expression in a subset of hemocytes, and that transgene expression is triggered after a blood meal. The hml promoter drives transgene expression in differentiating prohemocytes as well as in differentiated granulocytes. Analysis of different immune markers in hemocytes in which the hml promoter drives transgene expression revealed that this regulatory region could be used to study phagocytosis as well as melanization. Finally, the hml promoter drives transgene expression in hemocytes in which o'nyong-nyong virus replicates. Altogether, the Drosophila hml promoter constitutes a good tool to drive transgene expression in hemocyte only and to analyze the function of these cells and the genes they express during pathogen infection in Anopheles gambiae.
Collapse
Affiliation(s)
- Emilie Pondeville
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| | - Nicolas Puchot
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | | | - Guillaume Carissimo
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Mickael Poidevin
- Centre de Génétique Moléculaire, CNRS UPR 2167, Gif-sur-Yvette, France
| | - Robert M Waterhouse
- Department of Ecology and Evolution, Swiss Institute of Bioinformatics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Eric Marois
- CNRS UPR9022, INSERM U1257, Université de Strasbourg, Strasbourg, France
| | - Catherine Bourgouin
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| |
Collapse
|
34
|
Taking Insect Immunity to the Single-Cell Level. Trends Immunol 2020; 41:190-199. [DOI: 10.1016/j.it.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/11/2020] [Accepted: 01/12/2020] [Indexed: 12/16/2022]
|
35
|
King JG. Developmental and comparative perspectives on mosquito immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103458. [PMID: 31377103 DOI: 10.1016/j.dci.2019.103458] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Diseases spread by mosquitoes have killed more people than those spread by any other group of arthropod vectors and remain an important factor in determining global health and economic stability. The mosquito innate immune system can act to either modulate infection with human pathogens or fight off entomopathogens and increase the fitness and longevity of infected mosquitoes. While work remains towards understanding the larval immune system and the development of the mosquito immune system, it has recently become clearer that environmental factors heavily shape the developing mosquito immune system and continue to influence the adult immune system as well. The adult immune system has been well-studied and is known to involve multiple tissues and diverse molecular mechanisms. This review summarizes and synthesizes what is currently understood about the development of the mosquito immune system and includes comparisons of immune components unique to mosquitoes among the blood-feeding arthropods as well as important distinguishing factors between the anopheline and culicine mosquitoes. An explanation is included for how mosquito immunity factors into vector competence and vectorial capacity is presented along with a model for the interrelationships between nutrition, microbiome, pathogen interactions and behavior as they relate to mosquito development, immune status, adult female fitness and ultimately, vectorial capacity. Novel discoveries in the fields of mosquito ecoimmunology, neuroimmunology, and intracellular antiviral responses are highlighted.
Collapse
Affiliation(s)
- Jonas G King
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, 32 Creelman Street, Dorman 402, Mississippi State, MS 39762, USA.
| |
Collapse
|
36
|
Inhibitors of Eicosanoid Biosynthesis Reveal that Multiple Lipid Signaling Pathways Influence Malaria Parasite Survival in Anopheles gambiae. INSECTS 2019; 10:insects10100307. [PMID: 31547026 PMCID: PMC6835628 DOI: 10.3390/insects10100307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022]
Abstract
Eicosanoids are bioactive signaling lipids derived from the oxidation of fatty acids that act as important regulators of immune homeostasis and inflammation. As a result, effective anti-inflammatory drugs have been widely used to reduce pain and inflammation which target key eicosanoid biosynthesis enzymes. Conserved from vertebrates to insects, the use of these eicosanoid pathway inhibitors offer opportunities to evaluate the roles of eicosanoids in less-characterized insect systems. In this study, we examine the potential roles of eicosanoids on malaria parasite survival in the mosquito Anopheles gambiae. Using Plasmodium oocyst numbers to evaluate parasite infection, general or specific inhibitors of eicosanoid biosynthesis pathways were evaluated. Following the administration of dexamethasone and indomethacin, respective inhibitors of phospholipid A2 (PLA2) and cyclooxygenase (COX), oocyst numbers were unaffected. However, inhibition of lipoxygenase (LOX) activity through the use of esculetin significantly increased oocyst survival. In contrast, 12-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]-dodecanoic acid (AUDA), an inhibitor of epoxide hydroxylase (EH), decreased oocyst numbers. These experiments were further validated through RNAi experiments to silence candidate genes homologous to EH in An. gambiae to confirm their contributions to Plasmodium development. Similar to the results of AUDA treatment, the silencing of EH significantly reduced oocyst numbers. These results imply that specific eicosanoids in An. gambiae can have either agonist or antagonistic roles on malaria parasite survival in the mosquito host.
Collapse
|
37
|
Wang J, Yang B, Wang W, Song X, Jiang Q, Qiu L, Wang L, Song L. The Enhanced Immune Protection in Chinese Mitten Crab Eriocheir sinensis Against the Second Exposure to Bacteria Aeromonas hydrophila. Front Immunol 2019; 10:2041. [PMID: 31555272 PMCID: PMC6722218 DOI: 10.3389/fimmu.2019.02041] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidences suggest that the enhanced immune responses and increased protection against bacteria-induced mortality can be initiated after the primary exposure to various microbial communities and their components in various organisms including commercially valuable crustaceans. In the present study, the survival rate and immune responses of Chinese mitten crab Eriocheir sinensis were determined after an immune priming (IP) with formalin-killed Aeromonas hydrophila and an immune challenge (ICH) with the same but live pathogen (Ah group). A group in which the animals received a salt injection prior to challenge was maintained as control (Ns group). In the present study, it was shown that an IP with killed A. hydrophila can significantly protect the crabs against the ICH with a lethal dose of the live pathogen. The increased survival was associated with elevated rate and duration of phagocytosis. The antibacterial activity of the serum was significantly increased in Ah group compared to that in Ns group. Significant changes of phenoloxidase (PO) activities were also found between Ah and Ns group but not in Ah group between IP and ICH. No significant changes of lysozyme were found in Ah and NS group during the whole experiment except 3 h after IP. In addition, the levels of transcripts and protein of Dscam were increased in hemocytes of the crabs from Ah group. All the results suggested that a primary immune priming with a particular killed pathogen could induce an enhanced immunity in crabs when they were encountered secondly with the same live pathogen. The evidences of elevated immune protections in crabs would contribute to better understand the mechanism of immune priming in invertebrates.
Collapse
Affiliation(s)
- Jingjing Wang
- Marine Science and Engineering College, Qingdao Agricultural University, Qingdao, China.,Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Bin Yang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
| | - Xiaorui Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
| | - Qiufen Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Lingling Wang
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, China
| | - Linsheng Song
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.,Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, China
| |
Collapse
|
38
|
Caragata EP, Tikhe CV, Dimopoulos G. Curious entanglements: interactions between mosquitoes, their microbiota, and arboviruses. Curr Opin Virol 2019; 37:26-36. [PMID: 31176069 PMCID: PMC6768729 DOI: 10.1016/j.coviro.2019.05.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 11/22/2022]
Abstract
Mosquitoes naturally harbor a diverse community of microorganisms that play a crucial role in their biology. Mosquito-microbiota interactions are abundant and complex. They can dramatically alter the mosquito immune response, and impede or enhance a mosquito's ability to transmit medically important arboviral pathogens. Yet critically, given the massive public health impact of arboviral disease, few such interactions have been well characterized. In this review, we describe the current state of knowledge of the role of microorganisms in mosquito biology, how microbial-induced changes to mosquito immunity moderate infection with arboviruses, cases of mosquito-microbial-virus interactions with a defined mechanism, and the molecular interactions that underlie the endosymbiotic bacterium Wolbachia's ability to block virus infection in mosquitoes.
Collapse
Affiliation(s)
- Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Chinmay V Tikhe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
39
|
Barletta ABF, Trisnadi N, Ramirez JL, Barillas-Mury C. Mosquito Midgut Prostaglandin Release Establishes Systemic Immune Priming. iScience 2019; 19:54-62. [PMID: 31351392 PMCID: PMC6661395 DOI: 10.1016/j.isci.2019.07.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/10/2019] [Accepted: 07/08/2019] [Indexed: 12/22/2022] Open
Abstract
Anopheles gambiae mosquitoes that have been infected with Plasmodium mount a more effective immune response to a subsequent infection. Priming is established when Plasmodium invasion of the mosquito midgut allows contact of the gut microbiota with epithelial cells. This event is followed by a systemic release of a hemocyte differentiation factor (HDF) consisting of Lipoxin A4 bound to Evokin, a lipocalin carrier, which increases the proportion of circulating hemocytes. We show that mosquito midgut cells produce and release prostaglandin E2 (PGE2), which attracts hemocytes to the midgut surface and enhances their patrolling activity. Systemic injection of prostaglandins (PGs) recapitulates the priming response and enhances antiplasmodial immunity by triggering HDF production. Although insects lack cyclooxygenases, two heme peroxidases, HPX7 and HPX8, catalyze essential steps in PG biosynthesis in mosquitoes. Mosquito midgut PGE2 release attracts hemocytes and establishes a long-lasting enhanced systemic cellular immune response to Plasmodium infection. Plasmodium invasion or bacterial exposure triggers midgut prostaglandin synthesis Prostaglandins attract mosquito hemocytes and increase their patrolling activity Two midgut peroxidases, HPX7 and HPX8, catalyze midgut prostaglandin synthesis Systemic release of midgut prostaglandins is essential to establish immune priming
Collapse
Affiliation(s)
- Ana Beatriz F Barletta
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Nathanie Trisnadi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jose Luis Ramirez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
40
|
Chemical depletion of phagocytic immune cells in Anopheles gambiae reveals dual roles of mosquito hemocytes in anti- Plasmodium immunity. Proc Natl Acad Sci U S A 2019; 116:14119-14128. [PMID: 31235594 DOI: 10.1073/pnas.1900147116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mosquito immunity is composed of both cellular and humoral factors that provide protection from invading pathogens. Immune cells known as hemocytes, have been intricately associated with phagocytosis and innate immune signaling. However, the lack of genetic tools has limited hemocyte study despite their importance in mosquito anti-Plasmodium immunity. To address these limitations, we employ the use of a chemical-based treatment to deplete phagocytic immune cells in Anopheles gambiae, demonstrating the role of phagocytes in complement recognition and prophenoloxidase production that limit the ookinete and oocyst stages of malaria parasite development, respectively. Through these experiments, we also define specific subtypes of phagocytic immune cells in An. gambiae, providing insights beyond the morphological characteristics that traditionally define mosquito hemocyte populations. Together, this study represents a significant advancement in our understanding of the roles of mosquito phagocytes in mosquito vector competence and demonstrates the utility of clodronate liposomes as an important tool in the study of invertebrate immunity.
Collapse
|
41
|
Shaw DK, Tate AT, Schneider DS, Levashina EA, Kagan JC, Pal U, Fikrig E, Pedra JHF. Vector Immunity and Evolutionary Ecology: The Harmonious Dissonance. Trends Immunol 2018; 39:862-873. [PMID: 30301592 PMCID: PMC6218297 DOI: 10.1016/j.it.2018.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022]
Abstract
Recent scientific breakthroughs have significantly expanded our understanding of arthropod vector immunity. Insights in the laboratory have demonstrated how the immune system provides resistance to infection, and in what manner innate defenses protect against a microbial assault. Less understood, however, is the effect of biotic and abiotic factors on microbial-vector interactions and the impact of the immune system on arthropod populations in nature. Furthermore, the influence of genetic plasticity on the immune response against vector-borne pathogens remains mostly elusive. Herein, we discuss evolutionary forces that shape arthropod vector immunity. We focus on resistance, pathogenicity and tolerance to infection. We posit that novel scientific paradigms should emerge when molecular immunologists and evolutionary ecologists work together.
Collapse
Affiliation(s)
- Dana K Shaw
- Department of Veterinary Microbiology and Pathology, Washington State, Pullman, WA, USA.
| | - Ann T Tate
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| | - David S Schneider
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Elena A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jonathan C Kagan
- Division of Gastroenterology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
42
|
Melillo D, Marino R, Italiani P, Boraschi D. Innate Immune Memory in Invertebrate Metazoans: A Critical Appraisal. Front Immunol 2018; 9:1915. [PMID: 30186286 PMCID: PMC6113390 DOI: 10.3389/fimmu.2018.01915] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 08/02/2018] [Indexed: 12/31/2022] Open
Abstract
The ability of developing immunological memory, a characteristic feature of adaptive immunity, is clearly present also in innate immune responses. In fact, it is well known that plants and invertebrate metazoans, which only have an innate immune system, can mount a faster and more effective response upon re-exposure to a stimulus. Evidence of immune memory in invertebrates comes from studies in infection immunity, natural transplantation immunity, individual, and transgenerational immune priming. These studies strongly suggest that environment and lifestyle take part in the development of immunological memory. However, in several instances the formal correlation between the phenomenon of immune memory and molecular and functional immune parameters is still missing. In this review, we have critically examined the cellular and humoral aspects of the invertebrate immune memory responses. In particular, we have focused our analysis on studies that have addressed immune memory in the most restrictive meaning of the term, i.e., the response to a challenge of a quiescent immune system that has been primed in the past. These studies highlight the central role of an increase in the number of immune cells and of their epigenetic re-programming in the establishment of sensu stricto immune memory in invertebrates.
Collapse
Affiliation(s)
- Daniela Melillo
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Naples, Italy
| | - Rita Marino
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Paola Italiani
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Naples, Italy
| | - Diana Boraschi
- Institute of Protein Biochemistry (IBP), National Research Council (CNR), Naples, Italy.,Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| |
Collapse
|
43
|
Futo M, Sell MP, Kutzer MAM, Kurtz J. Specificity of oral immune priming in the red flour beetle Tribolium castaneum. Biol Lett 2018; 13:rsbl.2017.0632. [PMID: 29237813 DOI: 10.1098/rsbl.2017.0632] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022] Open
Abstract
Immune specificity is the degree to which a host's immune system discriminates among various pathogens or antigenic variants. Vertebrate immune memory is highly specific due to antibody responses. On the other hand, some invertebrates show immune priming, i.e. improved survival after secondary exposure to a previously encountered pathogen. Until now, specificity of priming has only been demonstrated via the septic infection route or when live pathogens were used for priming. Therefore, we tested for specificity in the oral priming route in the red flour beetle, Tribolium castaneum For priming, we used pathogen-free supernatants derived from three different strains of the entomopathogen, Bacillus thuringiensis, which express different Cry toxin variants known for their toxicity against this beetle. Subsequent exposure to the infective spores showed that oral priming was specific for two naturally occurring strains, while a third engineered strain did not induce any priming effect. Our data demonstrate that oral immune priming with a non-infectious bacterial agent can be specific, but the priming effect is not universal across all bacterial strains.
Collapse
Affiliation(s)
- Momir Futo
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany
| | - Marie P Sell
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany
| | - Megan A M Kutzer
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany
| | - Joachim Kurtz
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany
| |
Collapse
|
44
|
Lv Z, Qiu L, Wang M, Jia Z, Wang W, Xin L, Liu Z, Wang L, Song L. Comparative study of three C1q domain containing proteins from pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 78:42-51. [PMID: 28923592 DOI: 10.1016/j.dci.2017.09.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 06/23/2017] [Accepted: 09/14/2017] [Indexed: 06/07/2023]
Abstract
C1q domain containing proteins (C1qDCs) are a family of proteins containing a globular head C1q domain (ghC1q) in C-terminus, which serve as pattern recognition receptors (PRRs) and mediate a series of immune responses. In the present study, three C1qDC proteins from pacific oyster Crassostrea gigas (CgC1qDC-2, CgC1qDC-3, CgC1qDC-4) were characterized and comparatively investigated to understand their roles in the immune response. All the three recombinant CgC1qDC proteins (rCgC1qDCs) could bind lipopolysaccharide (LPS) significantly but they could not bind lipoteichoic acid (LTA), β-1,3-glucan (GLU), mannan (MAN), and polyinosinic-polycytidylic acid (Poly I:C). Correspondingly, they all exhibited higher binding activities towards Gram-negative bacteria Vibrio anguillarum and V. splendidus. Moreover, they could enhance the phagocytosis of oyster hemocytes, and the enhancements towards Gram-negative bacteria were significantly higher than that towards Gram-positive bacteria (p < 0.01). The LPS binding affinity of rCgC1qDC-3 (KD = 8.74 × 10-7 M) was higher than that of rCgC1qDC-2 (KD = 7.76 × 10-5 M) and rCgC1qDC-4 (KD = 1.09 × 10-5 M). Meanwhile, rCgC1qDC-3 exhibited significantly higher enhancement on phagocytosis of oyster hemocytes towards Gram-negative bacteria than that of rCgC1qDC-2 and rCgC1qDC-4 (p < 0.05). After the secondary challenge with V. splendidus, the up-regulations of CgC1qDC-2 and CgC1qDC-4 mRNA in hemocytes occurred at 6 h, while that of CgC1qDC-3 was observed at 3 h and lasted for 24 h. And CgC1qDC-3 responded with high mRNA level for tested 24 h upon the secondary challenge with V. anguillarum as well. These results collectively suggested that three CgC1qDCs could serve as PRRs to specifically recognize certain Gram-negative bacteria and opsonins to enhance phagocytosis. CgC1qDC-3, with higher binding affinity to LPS, stronger opsonization and more rapid and persistent mRNA expression response upon the secondary challenge with homologous Vibrios, might exert efficient functions in the immune responses against invading pathogens.
Collapse
Affiliation(s)
- Zhao Lv
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Mengqiang Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Zhihao Jia
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weilin Wang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lusheng Xin
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaoqun Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingling Wang
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
45
|
Kwon H, Arends BR, Smith RC. Late-phase immune responses limiting oocyst survival are independent of TEP1 function yet display strain specific differences in Anopheles gambiae. Parasit Vectors 2017; 10:369. [PMID: 28764765 PMCID: PMC5540282 DOI: 10.1186/s13071-017-2308-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/25/2017] [Indexed: 11/18/2022] Open
Abstract
Background There is emerging evidence that mosquito anti-Plasmodium immunity is multimodal with distinct mechanisms for killing malaria parasites at either the ookinete or oocyst stages. Early-phase responses targeting the ookinete require complement-like components circulating in the mosquito hemolymph that result in TEP1-mediated lysis or melanization. Additional responses mediated by the LL3 and STAT pathways limit oocyst survival through unknown mechanisms that require mosquito hemocyte function. While previous experiments argue that these mechanisms of parasite killing are independent, the transient nature of gene-silencing has rendered these experiments inconclusive. To address this issue, we outline experiments using a TALEN-derived TEP1 mutant line to examine the role of TEP1 in the Anopheles gambiae late-phase immune response. Results Despite higher early oocyst numbers in the TEP1 mutant line, no differences in oocyst survival were observed when compared to control mosquitoes, suggesting that TEP1 function is independent of the late-phase immune response. To further validate this phenotype in the TEP1 mutant, oocyst survival was evaluated in the TEP1 mutant background by silencing either LL3 or STAT-A. Surprisingly, only STAT-A silenced mosquitoes were able to reconstitute the late-phase immune phenotype increasing oocyst survival in the TEP1 mutant line. Additional experiments highlight significant differences in LL3 expression in the M/S hybrid genetic background of the TEP1 mutant line compared to that of the Keele strain (M form) of An. gambiae, and demonstrate that LL3 is not required for granulocyte differentiation in the M/S hybrid G3 genetic background in response to malaria parasite infection. Conclusions Through the combination of genetic experiments utilizing genetic mutants and reverse genetic approaches, new information has emerged regarding the mechanisms of mosquito late-phase immunity. When combined with previously published experiments, the body of evidence argues that Plasmodium oocyst survival is TEP1 independent, thus establishing that the mechanisms of early- and late-phase immunity are distinct. Moreover, we identify that the known components that mediate oocyst survival are susceptible to strain-specific differences depending on their genetic background and provide further evidence that the signals that promote hemocyte differentiation are required to limit oocyst survival. Together, this study provides new insights into the mechanisms of oocyst killing and the importance of genetics in shaping mosquito vector competence. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-2308-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA
| | - Benjamin R Arends
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
46
|
Kim IH, Pham V, Jablonka W, Goodman WG, Ribeiro JMC, Andersen JF. A mosquito hemolymph odorant-binding protein family member specifically binds juvenile hormone. J Biol Chem 2017; 292:15329-15339. [PMID: 28751377 DOI: 10.1074/jbc.m117.802009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/20/2017] [Indexed: 11/06/2022] Open
Abstract
Juvenile hormone (JH) is a key regulator of insect development and reproduction. In adult mosquitoes, it is essential for maturation of the ovary and normal male reproductive behavior, but how JH distribution and activity is regulated after secretion is unclear. Here, we report a new type of specific JH-binding protein, given the name mosquito juvenile hormone-binding protein (mJHBP), which circulates in the hemolymph of pupal and adult Aedes aegypti males and females. mJHBP is a member of the odorant-binding protein (OBP) family, and orthologs are present in the genomes of Aedes, Culex, and Anopheles mosquito species. Using isothermal titration calorimetry, we show that mJHBP specifically binds JH II and JH III but not eicosanoids or JH derivatives. mJHBP was crystallized in the presence of JH III and found to have a double OBP domain structure reminiscent of salivary "long" D7 proteins of mosquitoes. We observed that a single JH III molecule is contained in the N-terminal domain binding pocket that is closed in an apparent conformational change by a C-terminal domain-derived α-helix. The electron density for the ligand indicated a high occupancy of the natural 10R enantiomer of JH III. Of note, mJHBP is structurally unrelated to hemolymph JHBP from lepidopteran insects. A low level of expression of mJHBP in Ae. aegypti larvae suggests that it is primarily active during the adult stage where it could potentially influence the effects of JH on egg development, mating behavior, feeding, or other processes.
Collapse
Affiliation(s)
- Il Hwan Kim
- From the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 and
| | - Van Pham
- From the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 and
| | - Willy Jablonka
- From the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 and
| | - Walter G Goodman
- the Department of Entomology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - José M C Ribeiro
- From the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 and
| | - John F Andersen
- From the Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland 20852 and .,the Department of Entomology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
47
|
Greenwood JM, Milutinović B, Peuß R, Behrens S, Esser D, Rosenstiel P, Schulenburg H, Kurtz J. Oral immune priming with Bacillus thuringiensis induces a shift in the gene expression of Tribolium castaneum larvae. BMC Genomics 2017; 18:329. [PMID: 28446171 PMCID: PMC5405463 DOI: 10.1186/s12864-017-3705-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 04/12/2017] [Indexed: 12/22/2022] Open
Abstract
Background The phenomenon of immune priming, i.e. enhanced protection following a secondary exposure to a pathogen, has now been demonstrated in a wide range of invertebrate species. Despite accumulating phenotypic evidence, knowledge of its mechanistic underpinnings is currently very limited. Here we used the system of the red flour beetle, Tribolium castaneum and the insect pathogen Bacillus thuringiensis (Bt) to further our molecular understanding of the oral immune priming phenomenon. We addressed how ingestion of bacterial cues (derived from spore supernatants) of an orally pathogenic and non-pathogenic Bt strain affects gene expression upon later challenge exposure, using a whole-transcriptome sequencing approach. Results Whereas gene expression of individuals primed with the orally non-pathogenic strain showed minor changes to controls, we found that priming with the pathogenic strain induced regulation of a large set of distinct genes, many of which are known immune candidates. Intriguingly, the immune repertoire activated upon priming and subsequent challenge qualitatively differed from the one mounted upon infection with Bt without previous priming. Moreover, a large subset of priming-specific genes showed an inverse regulation compared to their regulation upon challenge only. Conclusions Our data demonstrate that gene expression upon infection is strongly affected by previous immune priming. We hypothesise that this shift in gene expression indicates activation of a more targeted and efficient response towards a previously encountered pathogen, in anticipation of potential secondary encounter. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3705-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jenny M Greenwood
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany
| | - Barbara Milutinović
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany.,Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
| | - Robert Peuß
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany.,Current Address: Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO, 64110, USA
| | - Sarah Behrens
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany
| | - Daniela Esser
- Institute of Clinical Molecular Biology, Christian-Albrechts University Kiel, Schittenhelmstr. 12, 24105, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts University Kiel, Schittenhelmstr. 12, 24105, Kiel, Germany
| | - Hinrich Schulenburg
- Zoological Institute, Christian-Albrechts University Kiel, Am Botanischen Garten 1-9, 24118, Kiel, Germany
| | - Joachim Kurtz
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany.
| |
Collapse
|
48
|
Dalli J. Does promoting resolution instead of inhibiting inflammation represent the new paradigm in treating infections? Mol Aspects Med 2017; 58:12-20. [PMID: 28365269 DOI: 10.1016/j.mam.2017.03.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/19/2022]
Abstract
Infections arise when the host response is overwhelmed by pathogens leading to organ dysfunction. In some instances patients progress to more severe conditions, including septic shock, that are associated with increased mortality. Current strategies in treating infections aim at either blocking inflammation using inhibitors to pro-inflammatory molecules and/or inhibiting bacterial growth using antibiotics. These approaches find their origins in studies conducted by Joseph Lister who demonstrated that applying carbolic acid to wounds promoted wound healing without suppuration, reducing both the necessity of amputation and mortality. While this approach is still applicable to certain infections, inhibition of the immune response is also associated with increased mortality, especially in septic patients. In many instances sepsis survivors succumb later to persistent, recurrent, nosocomial and secondary infections. This, together with a rise in resistance to many frontline antibiotics, has prompted a search for alternative ways to treat infections. Recent studies investigating processes engaged by the host response during self-resolving infections identified a novel group of mediators, termed as specialized pro-resolving mediators (SPM). These molecules, produced via the enzymatic conversion of essential fatty acids, actively reprogram the immune response to promote clearance of invading pathogens, and counter-regulate the production of inflammation-initiating molecules. Furthermore, recent studies also demonstrate that these mediators promote tissue repair and regeneration, essential processes in the re-establishment of barrier and prevention of re-infection. The scope of the present review is to discuss the evidence underpinning the endogenous protective roles of these novel mediators, as well as the evidence demonstrating that dysregulation in their production and actions contribute to disease pathogenesis in infections. This review will also discuss the potential of resolution pharmacology-based approaches in developing new therapeutics for combatting infections that do not interfere with the immune response.
Collapse
Affiliation(s)
- Jesmond Dalli
- Lipid Mediator Unit, Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom.
| |
Collapse
|
49
|
Tate AT, Andolfatto P, Demuth JP, Graham AL. The within-host dynamics of infection in trans-generationally primed flour beetles. Mol Ecol 2017; 26:3794-3807. [PMID: 28277618 DOI: 10.1111/mec.14088] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 12/19/2022]
Abstract
Many taxa exhibit plastic immune responses initiated after primary microbial exposure that provide increased protection against disease-induced mortality and the fitness costs of infection. In several arthropod species, this protection can even be passed from parents to offspring through a phenomenon called trans-generational immune priming. Here, we first demonstrate that trans-generational priming is a repeatable phenomenon in flour beetles (Tribolium castaneum) primed and infected with Bacillus thuringiensis (Bt). We then quantify the within-host dynamics of microbes and host physiological responses in infected offspring from primed and unprimed mothers by monitoring bacterial density and using mRNA-seq to profile host gene expression, respectively, over the acute infection period. We find that priming increases inducible resistance against Bt around a critical temporal juncture where host septicaemic trajectories, and consequently survival, may be determined in unprimed individuals. Our results identify a highly differentially expressed biomarker of priming, containing an EIF4-e domain, in uninfected individuals, as well as several other candidate genes. Moreover, the induction and decay dynamics of gene expression over time suggest a metabolic shift in primed individuals. The identified bacterial and gene expression dynamics are likely to influence patterns of bacterial fitness and disease transmission in natural populations.
Collapse
Affiliation(s)
- Ann T Tate
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Peter Andolfatto
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Jeffery P Demuth
- Department of Biology, University of Texas, Arlington, TX, 76010, USA
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, 08544, USA
| |
Collapse
|
50
|
Sadekuzzaman M, Park Y, Lee S, Kim K, Jung JK, Kim Y. An entomopathogenic bacterium, Xenorhabdus hominickii ANU101, produces oxindole and suppresses host insect immune response by inhibiting eicosanoid biosynthesis. J Invertebr Pathol 2017; 145:13-22. [PMID: 28302381 DOI: 10.1016/j.jip.2017.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/06/2017] [Accepted: 03/10/2017] [Indexed: 11/16/2022]
Abstract
An entomopathogenic bacterium, Xenorhabdus hominickii ANU101, was isolated from an entomopathogenic nematode, Steinernema monticolum. X. hominickii exhibited significant insecticidal activities at ≥6.6×102 colony-forming units per larva against a lepidopteran insect, Spodoptera exigua with hemocoelic injection. The insecticidal activity of X. hominickii was reduced by an addition of arachidonic acid (AA, a catalytic product of PLA2), but enhanced by an addition by dexamethasone (DEX, a specific inhibitor of PLA2). S. exigua could defend the bacterial infection by forming hemocyte nodules. However, live X. hominickii significantly reduced the hemocytic nodulation compared to similar treatment with heat-killed X. hominickii. An addition of AA to live X. hominickii significantly rescued the immunosuppression. X. hominickii also inhibited phenoloxidase activity in hemolymph of S. exigua larvae. Furthermore, the bacteria suppressed gene expressions of antimicrobial peptides, such as attacin-1, attacin-2, defensin, gallerimycin and transferrin-1 of S. exigua. An organic extract of X. hominickii-cultured broth with ethyl acetate possessed oxindole and significantly suppressed hemocyte nodulation. Again, an addition of AA diminished the inhibitory activity of the organic extract against hemocyte nodulation. Oxindole alone inhibited hemocyte nodulation and PLA2 enzyme activity. These results suggest that the entomopathogenicity of X. hominickii comes from its inhibitory activity against eicosanoid biosynthesis of target insects.
Collapse
Affiliation(s)
- Md Sadekuzzaman
- Department of Bioresource Sciences, Andong National University, Andong 36729, Republic of Korea
| | - Youngjin Park
- Department of Bioresource Sciences, Andong National University, Andong 36729, Republic of Korea
| | - Seunghee Lee
- Department of Bioresource Sciences, Andong National University, Andong 36729, Republic of Korea
| | - Kunwoo Kim
- Department of Bioresource Sciences, Andong National University, Andong 36729, Republic of Korea
| | - Jin Kyo Jung
- Division of Crop Cultivation and Environment Research, Department of Central Area Crop Science, National Institute of Crop Science, Rural Development Administration, Suwon 16429, Republic of Korea
| | - Yonggyun Kim
- Department of Bioresource Sciences, Andong National University, Andong 36729, Republic of Korea.
| |
Collapse
|