1
|
Böhmer P, Reuter M, Altinli M, Schnettler E. Detection and Characterization of Antiviral Proteins Against Bunyaviruses in Mosquito-Derived Cells. Methods Mol Biol 2025; 2893:73-84. [PMID: 39671031 DOI: 10.1007/978-1-0716-4338-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Viruses of the class Bunyaviricetes are often transmitted by arthropods, including mosquitoes. The innate immune response in mosquitoes comprises several pathways, including sequence-specific degradation through RNA interference (RNAi). It is known that bunyavirus infections are targeted by the innate immune response in mosquitoes and derived cells; however, detailed information is often still missing. In this chapter, we describe a detailed protocol to determine if a protein of interest acts antiviral against bunyavirus infection in mosquito-derived cells. In line, we also describe a detailed protocol to investigate the effect of overexpression of a mosquito-encoded protein or analysis of mutant proteins on bunyavirus infection.
Collapse
Affiliation(s)
- P Böhmer
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - M Reuter
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - M Altinli
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - E Schnettler
- Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany.
- Faculty of Mathematics, Informatics and Natural Sciences, University Hamburg, Hamburg, Germany.
| |
Collapse
|
2
|
Hu R, Li M, Chen S, Wang M, Tao X, Zhu Y, Yan H, Liu Y. Sniffer restricts arboviral brain infections by regulating ROS levels and protecting blood-brain barrier integrity in Drosophila and mosquitoes. PLoS Pathog 2024; 20:e1012797. [PMID: 39680616 DOI: 10.1371/journal.ppat.1012797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Arthropod-borne viruses (arboviruses) are transmitted to humans by arthropod vectors and pose a serious threat to global public health. Neurotropic arboviruses including Sindbis virus (SINV) persistently infect the central nervous system (CNS) of vector insects without causing notable pathological changes or affecting their behavior or lifespan. However, the mechanisms by which vector insects evade these viral infections in the brains are poorly understood. In this study, we found that loss of the carbonyl reductase Sniffer (Sni) led to a significant increase in SINV infection in the Drosophila brain. Sni regulates reactive oxygen species (ROS) levels, and its depletion leads to elevated ROS, which in turn disrupts the septate junctions (SJs) between subperineurial glia (SPG) cells, compromising the integrity and barrier function of the blood-brain barrier (BBB). Genetic and pharmacological reduction of ROS restored BBB integrity and reduced viral load in the brains of Sni-depleted flies. Additionally, we identified Sni homologs and revealed that the antiviral function of Sni is highly conserved in mosquitoes, where it regulates ROS and protects BBB integrity. Our results revealed an evolutionarily conserved antiviral mechanism in which Sni acts as an antioxidant that protects BBB integrity and restricts viral infection in the vector insect brain.
Collapse
Affiliation(s)
- Rui Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mengzhu Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shulin Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Man Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xinjun Tao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yihan Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huan Yan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yuan Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
vom Hemdt A, Thienel AL, Ciupka K, Wieseler J, Proksch HM, Schlee M, Kümmerer BM. 2'-O-methyltransferase-deficient yellow fever virus: Restricted replication in the midgut and secondary tissues of Aedes aegypti mosquitoes severely limits dissemination. PLoS Pathog 2024; 20:e1012607. [PMID: 39356716 PMCID: PMC11472933 DOI: 10.1371/journal.ppat.1012607] [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: 04/09/2024] [Revised: 10/14/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The RNA genome of orthoflaviviruses encodes a methyltransferase within the non-structural protein NS5, which is involved in 2'-O-methylation of the 5'-terminal nucleotide of the viral genome resulting in a cap1 structure. While a 2'-O-unmethylated cap0 structure is recognized in vertebrates by the RNA sensor RIG-I, the cap1 structure allows orthoflaviviruses to evade the vertebrate innate immune system. Here, we analyzed whether the cap0 structure is also recognized in mosquitoes. Replication analyses of 2'-O-methyltransferase deficient yellow fever virus mutants (YFV NS5-E218A) of the vaccine 17D and the wild-type Asibi strain in mosquito cells revealed a distinct downregulation of the cap0 viruses. Interestingly, the level of inhibition differed for various mosquito cells. The most striking difference was found in Aedes albopictus-derived C6/36 cells with YFV-17D cap0 replication being completely blocked. Replication of YFV-Asibi cap0 was also suppressed in mosquito cells but to a lower extent. Analyses using chimeras between YFV-17D and YFV-Asibi suggest that a synergistic effect of several mutations across the viral genome accompanied by a faster initial growth rate of YFV-Asibi cap1 correlates with the lower level of YFV-Asibi cap0 attenuation. Viral growth analyses in Dicer-2 knockout cells demonstrated that Dicer-2 is entirely dispensable for attenuating the YFV cap0 viruses. Translation of a replication-incompetent cap0 reporter YFV-17D genome was reduced in mosquito cells, indicating a cap0 sensing translation regulation mechanism. Further, oral infection of Aedes aegypti mosquitoes resulted in lower infection rates for YFV-Asibi cap0. The latter is related to lower viral loads found in the midguts, which largely diminished dissemination to secondary tissues. After intrathoracic infection, YFV-Asibi cap0 replicated slower and to decreased amounts in secondary tissues compared to YFV-Asibi cap1. These results suggest the existence of an ubiquitously expressed innate antiviral protein recognizing 5'-terminal RNA cap-modifications in mosquitoes, both in the midgut as well as in secondary tissues.
Collapse
Affiliation(s)
- Anja vom Hemdt
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Katrin Ciupka
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Janett Wieseler
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Hannah M. Proksch
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Beate M. Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
4
|
Cui Y, Megawati D, Lin J, Rehard DG, Grant DG, Liu P, Jurkevich A, Reid WR, Mooney BP, Franz AW. Cytoskeleton-associated gelsolin responds to the midgut distention process in saline meal-fed Aedes aegypti and affects arbovirus dissemination from the midgut. FASEB J 2024; 38:e23764. [PMID: 39042395 PMCID: PMC11268798 DOI: 10.1096/fj.202302684rr] [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: 12/27/2023] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024]
Abstract
The mosquito, Aedes aegypti, is the principal vector for several arboviruses. The mosquito midgut is the initial tissue that gets infected with an arbovirus acquired along with a blood meal from a vertebrate host. Blood meal ingestion leads to midgut tissue distention thereby increasing the pore size of the surrounding basal lamina. This allows newly synthesized virions to exit the midgut by traversing the distended basal lamina to infect secondary tissues of the mosquito. We conducted a quantitative label-free proteomic time course analysis with saline meal-fed Ae. aegypti females to identify host factors involved in midgut tissue distention. Around 2000 proteins were detected during each of the seven sampling time points and 164 of those were uniquely expressed. Forty-five of 97 differentially expressed proteins were upregulated during the 96-h time course and most of those were involved in cytoskeleton modulation, metabolic activity, and vesicle/vacuole formation. The F-actin-modulating Ae. aegypti (Aa)-gelsolin was selected for further functional studies. Stable knockout of Aa-gelsolin resulted in a mosquito line, which showed distorted actin filaments in midgut-associated tissues likely due to diminished F-actin processing by gelsolin. Zika virus dissemination from the midgut of these mosquitoes was diminished and delayed. The loss of Aa-gelsolin function was associated with an increased induction of apoptosis in midgut tissue indicating an involvement of Aa-gelsolin in apoptotic signaling in mosquitoes. Here, we used proteomics to discover a novel host factor, Aa-gelsolin, which affects the midgut escape barrier for arboviruses in mosquitoes and apoptotic signaling in the midgut.
Collapse
Affiliation(s)
- Yingjun Cui
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Dewi Megawati
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
- Department of Microbiology and Parasitology, School of Medicine, Warmadewa University, Bali, Indonesia
| | - Jingyi Lin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - David G. Rehard
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - DeAna G. Grant
- Electron Microscopy Core, University of Missouri, Columbia, Missouri, USA
| | - Pei Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri, USA
| | - Alexander Jurkevich
- Advanced Light Microscopy Core, University of Missouri, Columbia, Missouri, USA
| | - William R. Reid
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Brian P. Mooney
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri, USA
| | - Alexander W.E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
5
|
Madhav M, Blasdell KR, Trewin B, Paradkar PN, López-Denman AJ. Culex-Transmitted Diseases: Mechanisms, Impact, and Future Control Strategies using Wolbachia. Viruses 2024; 16:1134. [PMID: 39066296 PMCID: PMC11281716 DOI: 10.3390/v16071134] [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: 06/12/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Mosquitoes of the Culex genus are responsible for a large burden of zoonotic virus transmission globally. Collectively, they play a significant role in the transmission of medically significant diseases such as Japanese encephalitis virus and West Nile virus. Climate change, global trade, habitat transformation and increased urbanisation are leading to the establishment of Culex mosquitoes in new geographical regions. These novel mosquito incursions are intensifying concerns about the emergence of Culex-transmitted diseases and outbreaks in previously unaffected areas. New mosquito control methods are currently being developed and deployed globally. Understanding the complex interaction between pathogens and mosquitoes is essential for developing new control strategies for Culex species mosquitoes. This article reviews the role of Culex mosquitos as vectors of zoonotic disease, discussing the transmission of viruses across different species, and the potential use of Wolbachia technologies to control disease spread. By leveraging the insights gained from recent successful field trials of Wolbachia against Aedes-borne diseases, we comprehensively discuss the feasibility of using this technique to control Culex mosquitoes and the potential for the development of next generational Wolbachia-based control methods.
Collapse
Affiliation(s)
- Mukund Madhav
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, VIC 3220, Australia
| | - Kim R. Blasdell
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, VIC 3220, Australia
| | - Brendan Trewin
- CSIRO Health and Biosecurity, Dutton Park, Brisbane, QLD 4102, Australia
| | - Prasad N. Paradkar
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, VIC 3220, Australia
| | - Adam J. López-Denman
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, VIC 3220, Australia
| |
Collapse
|
6
|
Feng T, Tong H, Zhang F, Zhang Q, Zhang H, Zhou X, Ruan H, Wu Q, Dai J. Transcriptome study reveals tick immune genes restrict Babesia microti infection. INSECT SCIENCE 2024. [PMID: 38837613 DOI: 10.1111/1744-7917.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Abstract
A systems biology approach was employed to gain insight into tick biology and interactions between vectors and pathogens. Haemaphysalis longicornis serves as one of the primary vectors of Babesia microti, significantly impacting human and animal health. Obtaining more information about their relationship is crucial for a comprehensive understanding of tick and pathogen biology, pathogen transmission dynamics, and potential control strategies. RNA sequencing of uninfected and B. microti-infected ticks resulted in the identification of 15 056 unigenes. Among these, 1 051 were found to be differentially expressed, with 796 being upregulated and 255 downregulated (P < 0.05). Integrated transcriptomics datasets revealed the pivotal role of immune-related pathways, including the Toll, Janus kinase/signal transducer and activator of transcription (JAK-STAT), immunodeficiency, and RNA interference (RNAi) pathways, in response to infection. Consequently, 3 genes encoding critical transcriptional factor Dorsal, Relish, and STAT were selected for RNAi experiments. The knockdown of Dorsal, Relish, and STAT resulted in a substantial increase in Babesia infection levels compared to the respective controls. These findings significantly advanced our understanding of tick-Babesia molecular interactions and proposed novel tick antigens as potential vaccine targets against tick infestations and pathogen transmission.
Collapse
Affiliation(s)
- Tingting Feng
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Tong
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Feihu Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Qianqian Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Heng Zhang
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xia Zhou
- School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Hang Ruan
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jianfeng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Bevivino G, Maurizi L, Ammendolia MG, Longhi C, Arcà B, Lombardo F. Peptides with Antimicrobial Activity in the Saliva of the Malaria Vector Anopheles coluzzii. Int J Mol Sci 2024; 25:5529. [PMID: 38791567 PMCID: PMC11121840 DOI: 10.3390/ijms25105529] [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: 04/27/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Mosquito saliva plays a crucial physiological role in both sugar and blood feeding by helping sugar digestion and exerting antihemostatic functions. During meal acquisition, mosquitoes are exposed to the internalization of external microbes. Since mosquitoes reingest significant amounts of saliva during feeding, we hypothesized that salivary antimicrobial components may participate in the protection of mouthparts, the crop, and the gut by inhibiting bacterial growth. To identify novel potential antimicrobials from mosquito saliva, we selected 11 candidates from Anopheles coluzzii salivary transcriptomic datasets and obtained them either using a cell-free transcription/translation expression system or, when feasible, via chemical synthesis. Hyp6.2 and hyp13, which were predicted to be produced as propeptides and cleaved in shorter mature forms, showed the most interesting results in bacterial growth inhibition assays. Hyp6.2 (putative mature form, 35 amino acid residues) significantly inhibited the growth of Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli and Serratia marcescens) bacteria. Hyp13 (short form, 19 amino acid residues) dose-dependently inhibited E. coli and S. marcescens growth, inducing membrane disruption in both Gram-positive and Gram-negative bacteria as indicated with scanning electron microscopy. In conclusion, we identified two A. coluzzii salivary peptides inhibiting Gram-positive and Gram-negative bacteria growth and possibly contributing to the protection of mosquito mouthparts and digestive tracts from microbial infection during and/or after feeding.
Collapse
Affiliation(s)
- Giulia Bevivino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Linda Maurizi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Maria Grazia Ammendolia
- National Center for Innovative Technologies in Public Health, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Catia Longhi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| |
Collapse
|
8
|
Imler JL, Cai H, Meignin C, Martins N. Evolutionary immunology to explore original antiviral strategies. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230068. [PMID: 38497262 PMCID: PMC10945398 DOI: 10.1098/rstb.2023.0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/25/2023] [Indexed: 03/19/2024] Open
Abstract
Over the past 25 years, the field of evolutionary developmental biology (evo-devo) has used genomics and genetics to gain insight on the developmental mechanisms underlying the evolution of morphological diversity of animals. Evo-devo exploits the key insight that conserved toolkits of development (e.g. Hox genes) are used in animals to produce genetic novelties that provide adaptation to a new environment. Like development, immunity is forged by interactions with the environment, namely the microbial world. Yet, when it comes to the study of immune defence mechanisms in invertebrates, interest primarily focuses on evolutionarily conserved molecules also present in humans. Here, focusing on antiviral immunity, we argue that immune genes not conserved in humans represent an unexplored resource for the discovery of new antiviral strategies. We review recent findings on the cGAS-STING pathway and explain how cyclic dinucleotides produced by cGAS-like receptors may be used to investigate the portfolio of antiviral genes in a broad range of species. This will set the stage for evo-immuno approaches, exploiting the investment in antiviral defences made by metazoans over hundreds of millions of years of evolution. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Jean-Luc Imler
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS UPR9022, Strasbourg 67070, France
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, People's Republic of China
| | - Hua Cai
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, People's Republic of China
| | - Carine Meignin
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS UPR9022, Strasbourg 67070, France
| | - Nelson Martins
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS UPR9022, Strasbourg 67070, France
| |
Collapse
|
9
|
Miranda LS, Rudd SR, Mena O, Hudspeth PE, Barboza-Corona JE, Park HW, Bideshi DK. The Perpetual Vector Mosquito Threat and Its Eco-Friendly Nemeses. BIOLOGY 2024; 13:182. [PMID: 38534451 DOI: 10.3390/biology13030182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Mosquitoes are the most notorious arthropod vectors of viral and parasitic diseases for which approximately half the world's population, ~4,000,000,000, is at risk. Integrated pest management programs (IPMPs) have achieved some success in mitigating the regional transmission and persistence of these diseases. However, as many vector-borne diseases remain pervasive, it is obvious that IPMP successes have not been absolute in eradicating the threat imposed by mosquitoes. Moreover, the expanding mosquito geographic ranges caused by factors related to climate change and globalization (travel, trade, and migration), and the evolution of resistance to synthetic pesticides, present ongoing challenges to reducing or eliminating the local and global burden of these diseases, especially in economically and medically disadvantaged societies. Abatement strategies include the control of vector populations with synthetic pesticides and eco-friendly technologies. These "green" technologies include SIT, IIT, RIDL, CRISPR/Cas9 gene drive, and biological control that specifically targets the aquatic larval stages of mosquitoes. Regarding the latter, the most effective continues to be the widespread use of Lysinibacillus sphaericus (Ls) and Bacillus thuringiensis subsp. israelensis (Bti). Here, we present a review of the health issues elicited by vector mosquitoes, control strategies, and lastly, focus on the biology of Ls and Bti, with an emphasis on the latter, to which no resistance has been observed in the field.
Collapse
Affiliation(s)
- Leticia Silva Miranda
- Graduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| | - Sarah Renee Rudd
- Graduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
- Integrated Biomedical Graduate Studies, and School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Oscar Mena
- Undergraduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| | - Piper Eden Hudspeth
- Undergraduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| | - José E Barboza-Corona
- Departmento de Alimentos, Posgrado en Biociencias, Universidad de Guanajuato Campus Irapuato-Salamanca, Irapuato 36500, Guanajuato, Mexico
| | - Hyun-Woo Park
- Graduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
- Undergraduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| | - Dennis Ken Bideshi
- Graduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
- Undergraduate Program in Biomedical Sciences, Department of Biological Sciences, California Baptist University, Riverside, CA 92504, USA
| |
Collapse
|
10
|
Zhang J, Tang T, Zhang R, Wen L, Deng X, Xu X, Yang W, Jin F, Cao Y, Lu Y, Yu XQ. Maintaining Toll signaling in Drosophila brain is required to sustain autophagy for dopamine neuron survival. iScience 2024; 27:108795. [PMID: 38292423 PMCID: PMC10825691 DOI: 10.1016/j.isci.2024.108795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 10/19/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Macroautophagy/autophagy is a conserved process in eukaryotic cells to degrade and recycle damaged intracellular components. Higher level of autophagy in the brain has been observed, and autophagy dysfunction has an impact on neuronal health, but the molecular mechanism is unclear. In this study, we showed that overexpression of Toll-1 and Toll-7 receptors, as well as active Spätzle proteins in Drosophila S2 cells enhanced autophagy, and Toll-1/Toll-7 activated autophagy was dependent on Tube-Pelle-PP2A. Interestingly, Toll-1 but not Toll-7 mediated autophagy was dMyd88 dependent. Importantly, we observed that loss of functions in Toll-1 and Toll-7 receptors and PP2A activity in flies decreased autophagy level, resulting in the loss of dopamine (DA) neurons and reduced fly motion. Our results indicated that proper activation of Toll-1 and Toll-7 pathways and PP2A activity in the brain are necessary to sustain autophagy level for DA neuron survival.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Ting Tang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ruonan Zhang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Liang Wen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiaojuan Deng
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoxia Xu
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Wanying Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Fengliang Jin
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou 510642, Guangdong, China
| | - Yang Cao
- Guangdong Laboratory for Lingnan Modern Agriculture, Laboratory of Insect Molecular Biology and Biotechnology, Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuzhen Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
11
|
Lewis J, Gallichotte EN, Randall J, Glass A, Foy BD, Ebel GD, Kading RC. Intrinsic factors driving mosquito vector competence and viral evolution: a review. Front Cell Infect Microbiol 2023; 13:1330600. [PMID: 38188633 PMCID: PMC10771300 DOI: 10.3389/fcimb.2023.1330600] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Mosquitoes are responsible for the transmission of numerous viruses of global health significance. The term "vector competence" describes the intrinsic ability of an arthropod vector to transmit an infectious agent. Prior to transmission, the mosquito itself presents a complex and hostile environment through which a virus must transit to ensure propagation and transmission to the next host. Viruses imbibed in an infectious blood meal must pass in and out of the mosquito midgut, traffic through the body cavity or hemocoel, invade the salivary glands, and be expelled with the saliva when the vector takes a subsequent blood meal. Viruses encounter physical, cellular, microbial, and immunological barriers, which are influenced by the genetic background of the mosquito vector as well as environmental conditions. Collectively, these factors place significant selective pressure on the virus that impact its evolution and transmission. Here, we provide an overview of the current state of the field in understanding the mosquito-specific factors that underpin vector competence and how each of these mechanisms may influence virus evolution.
Collapse
Affiliation(s)
- Juliette Lewis
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Emily N. Gallichotte
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Jenna Randall
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Arielle Glass
- Department of Cellular and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| | - Brian D. Foy
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Gregory D. Ebel
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Rebekah C. Kading
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
12
|
Rai P, Webb EM, Kang L, Weger-Lucarelli J. Insulin reduces the transmission potential of chikungunya virus and activates the toll pathway in Aedes aegypti mosquitoes. INSECT MOLECULAR BIOLOGY 2023; 32:648-657. [PMID: 37334906 DOI: 10.1111/imb.12863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
Chikungunya virus (CHIKV) is an alphavirus that has re-emerged globally over the last two decades and has the potential to become endemic in the United States due to the presence of competent mosquito vectors, Aedes aegypti and Aedes albopictus. CHIK disease is characterised by fever, rash, and joint pain, and causes chronic debilitating joint pain and swelling in >50% of infected individuals. Given the disease severity caused by CHIKV and the global presence of vectors to facilitate its spread, strategies to reduce viral transmission are desperately needed; however, the human biological factors driving CHIKV transmission are poorly understood. Towards that end, we have previously shown that mosquitoes fed on alphavirus-infected obese mice have reduced infection and transmission rates compared to those fed on infected lean mice despite similar viremia in lean and obese mice. One of the many host factors that increase in obese hosts is insulin, which was previously shown to impact the infection of mosquitoes by several flaviviruses. However, insulin's impact on alphavirus infection of live mosquitoes is unknown and whether insulin influences mosquito-borne virus transmission has not been tested. To test this, we exposed A. aegypti mosquitoes to bloodmeals with CHIKV in the presence or absence of physiologically relevant levels of insulin and found that insulin significantly lowered both infection and transmission rates. RNA sequencing analysis on mosquito midguts isolated at 1-day-post-infectious-bloodmeal (dpbm) showed enrichment in genes in the Toll immune pathway in the presence of insulin, which was validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We then sought to determine if the Toll pathway plays a role in CHIKV infection of Ae. aegypti mosquitoes; therefore, we knocked down Myd88, a critical immune adaptor molecule for the Toll pathway, in live mosquitoes, and found increased CHIKV infection compared to the mock knockdown control group. Overall, these data demonstrate that insulin reduces CHIKV transmission by Ae. aegypti and activates the Toll pathway in mosquitoes, suggesting that conditions resulting in higher serum insulin concentrations may reduce alphavirus transmission. Finally, these studies suggest that strategies to activate insulin or Toll signalling in mosquitoes may be an effective control strategy against medically relevant alphaviruses.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| | - Emily M Webb
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
- Department of Entomology, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
13
|
Hernandez-Caballero I, Hellgren O, Garcia-Longoria Batanete L. Genomic advances in the study of the mosquito vector during avian malaria infection. Parasitology 2023; 150:1330-1339. [PMID: 37614176 PMCID: PMC10941221 DOI: 10.1017/s0031182023000756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
Invertebrate host–parasite associations are one of the keystones in order to understand vector-borne diseases. The study of these specific interactions provides information not only about how the vector is affected by the parasite at the gene-expression level, but might also reveal mosquito strategies for blocking the transmission of the parasites. A very well-known vector for human malaria is Anopheles gambiae. This mosquito species has been the main focus for genomics studies determining essential key genes and pathways over the course of a malaria infection. However, to-date there is an important knowledge gap concerning other non-mammophilic mosquito species, for example some species from the Culex genera which may transmit avian malaria but also zoonotic pathogens such as West Nile virus. From an evolutionary perspective, these 2 mosquito genera diverged 170 million years ago, hence allowing studies in both species determining evolutionary conserved genes essential during malaria infections, which in turn might help to find key genes for blocking malaria cycle inside the mosquito. Here, we extensively review the current knowledge on key genes and pathways expressed in Anopheles over the course of malaria infections and highlight the importance of conducting genomic investigations for detecting pathways in Culex mosquitoes linked to infection of avian malaria. By pooling this information, we underline the need to increase genomic studies in mosquito–parasite associations, such as the one in Culex–Plasmodium, that can provide a better understanding of the infection dynamics in wildlife and reduce the negative impact on ecosystems.
Collapse
Affiliation(s)
- Irene Hernandez-Caballero
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, E-06071 Badajoz, Spain
| | - Olof Hellgren
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Sölvegatan 37, SE-22362, Sweden
| | | |
Collapse
|
14
|
Garrigós M, Garrido M, Panisse G, Veiga J, Martínez-de la Puente J. Interactions between West Nile Virus and the Microbiota of Culex pipiens Vectors: A Literature Review. Pathogens 2023; 12:1287. [PMID: 38003752 PMCID: PMC10675824 DOI: 10.3390/pathogens12111287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The flavivirus West Nile virus (WNV) naturally circulates between mosquitoes and birds, potentially affecting humans and horses. Different species of mosquitoes play a role as vectors of WNV, with those of the Culex pipiens complex being particularly crucial for its circulation. Different biotic and abiotic factors determine the capacity of mosquitoes for pathogen transmission, with the mosquito gut microbiota being recognized as an important one. Here, we review the published studies on the interactions between the microbiota of the Culex pipiens complex and WNV infections in mosquitoes. Most articles published so far studied the interactions between bacteria of the genus Wolbachia and WNV infections, obtaining variable results regarding the directionality of this relationship. In contrast, only a few studies investigate the role of the whole microbiome or other bacterial taxa in WNV infections. These studies suggest that bacteria of the genera Serratia and Enterobacter may enhance WNV development. Thus, due to the relevance of WNV in human and animal health and the important role of mosquitoes of the Cx. pipiens complex in its transmission, more research is needed to unravel the role of mosquito microbiota and those factors affecting this microbiota on pathogen epidemiology. In this respect, we finally propose future lines of research lines on this topic.
Collapse
Affiliation(s)
- Marta Garrigós
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Mario Garrido
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Guillermo Panisse
- CEPAVE—Centro de Estudios Parasitológicos y de Vectores CONICET-UNLP, La Plata 1900, Argentina;
| | - Jesús Veiga
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Josué Martínez-de la Puente
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
15
|
Cardoso-Jaime V, Maya-Maldonado K, Tsutsumi V, Hernández-Martínez S. Mosquito pericardial cells upregulate Cecropin expression after an immune challenge. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104745. [PMID: 37268262 DOI: 10.1016/j.dci.2023.104745] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Most mosquito-transmitted pathogens grow or replicate in the midgut before invading the salivary glands. Pathogens are exposed to several immunological factors along the way. Recently, it was shown that hemocytes gather near the periostial region of the heart to efficiently phagocytose pathogens circulating in the hemolymph. Nerveless, not all pathogens can be phagocyted by hemocytes and eliminated by lysis. Interestingly, some studies have shown that pericardial cells (PCs) surrounding periostial regions, may produce humoral factors, such as lysozymes. Our current work provides evidence that Anopheles albimanus PCs are a major producer of Cecropin 1 (Cec1). Furthermore, our findings reveal that after an immunological challenge, PCs upregulate Cec1 expression. We conclude that PCs are positioned in a strategic location that could allow releasing humoral components, such as cecropin, to lyse pathogens on the heart or circulating in the hemolymph, implying that PCs could play a significant role in the systemic immune response.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Krystal Maya-Maldonado
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Víctor Tsutsumi
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico.
| | - Salvador Hernández-Martínez
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico.
| |
Collapse
|
16
|
Abstract
Haematophagous arthropods, including mosquitoes, ticks, flies, triatomine bugs and lice (here referred to as vectors), are involved in the transmission of various pathogens to mammals on whom they blood feed. The diseases caused by these pathogens, collectively known as vector-borne diseases (VBDs), threaten the health of humans and animals. Although the vector arthropods differ in life histories, feeding behaviour as well as reproductive strategies, they all harbour symbiotic microorganisms, known as microbiota, on which they depend for completing essential aspects of their biology, such as development and reproduction. In this Review, we summarize the shared and unique key features of the symbiotic associations that have been characterized in the major vector taxa. We discuss the crosstalks between microbiota and their arthropod hosts that influence vector metabolism and immune responses relevant for pathogen transmission success, known as vector competence. Finally, we highlight how current knowledge on symbiotic associations is being explored to develop non-chemical-based alternative control methods that aim to reduce vector populations, or reduce vector competence. We conclude by highlighting the remaining knowledge gaps that stand to advance basic and translational aspects of vector-microbiota interactions.
Collapse
Affiliation(s)
- Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P. R. China.
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P. R. China.
| | - Li Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
17
|
Prince BC, Chan K, Rückert C. Elucidating the role of dsRNA sensing and Toll6 in antiviral responses of Culex quinquefasciatus cells. Front Cell Infect Microbiol 2023; 13:1251204. [PMID: 37712057 PMCID: PMC10499357 DOI: 10.3389/fcimb.2023.1251204] [Citation(s) in RCA: 1] [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: 07/01/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
The first step of any immune response is the recognition of foreign molecular structures inside the host organism. An important molecule that is generally foreign to eukaryotic cells is long double-stranded RNA (dsRNA), which can be generated during virus replication. The mechanisms of sensing viral dsRNA are well-studied in mammalian systems but are only poorly understood in insects, including disease vectors such as Culex quinquefasciatus mosquitoes. These mosquitoes are vectors for important arboviruses, such as West Nile virus, and Culex species mosquitoes are distributed across the globe in many temperate and tropical regions. The major antiviral response triggered by dsRNA in mosquitoes is RNA interference - a sequence-specific response which targets complementary viral RNA for degradation. However, here, we aimed to identify whether sequence-independent dsRNA sensing, mimicked by poly(I:C), can elicit an antiviral response. We observed a significant reduction in replication of La Crosse virus (LACV) in Cx. quinquefasciatus mosquito cells following poly(I:C) priming. We identified a number of antimicrobial peptides and Toll receptors that were upregulated at the transcript level by poly(I:C) stimulation. Notably, Toll6 was upregulated and we determined that a knockdown of Toll6 expression resulted also in increased LACV replication. Future efforts require genetic tools to validate whether the observed Toll6 antiviral activity is indeed linked to dsRNA sensing. However, large-scale functional genomic and proteomic approaches are also required to determine which downstream responses are part of the poly(I:C) elicited antiviral response.
Collapse
Affiliation(s)
| | | | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV, United States
| |
Collapse
|
18
|
Prince BC, Walsh E, Torres TZB, Rückert C. Recognition of Arboviruses by the Mosquito Immune System. Biomolecules 2023; 13:1159. [PMID: 37509194 PMCID: PMC10376960 DOI: 10.3390/biom13071159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to both human and animal health worldwide. These viruses are transmitted through the bites of mosquitoes, ticks, sandflies, or biting midges to humans or animals. In humans, arbovirus infection often results in mild flu-like symptoms, but severe disease and death also occur. There are few vaccines available, so control efforts focus on the mosquito population and virus transmission control. One area of research that may enable the development of new strategies to control arbovirus transmission is the field of vector immunology. Arthropod vectors, such as mosquitoes, have coevolved with arboviruses, resulting in a balance of virus replication and vector immune responses. If this balance were disrupted, virus transmission would likely be reduced, either through reduced replication, or even through enhanced replication, resulting in mosquito mortality. The first step in mounting any immune response is to recognize the presence of an invading pathogen. Recent research advances have been made to tease apart the mechanisms of arbovirus detection by mosquitoes. Here, we summarize what is known about arbovirus recognition by the mosquito immune system, try to generate a comprehensive picture, and highlight where there are still gaps in our current understanding.
Collapse
Affiliation(s)
- Brian C Prince
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Elizabeth Walsh
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Tran Zen B Torres
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
19
|
Belavilas-Trovas A, Tastsoglou S, Dong S, Kefi M, Tavadia M, Mathiopoulos KD, Dimopoulos G. Long non-coding RNAs regulate Aedes aegypti vector competence for Zika virus and reproduction. PLoS Pathog 2023; 19:e1011440. [PMID: 37319296 DOI: 10.1371/journal.ppat.1011440] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play critical regulatory roles in various cellular and metabolic processes in mosquitoes and all other organisms studied thus far. In particular, their involvement in essential processes such as reproduction makes them potential targets for the development of novel pest control approaches. However, their function in mosquito biology remains largely unexplored. To elucidate the role of lncRNAs in mosquitoes' reproduction and vector competence for arboviruses, we have implemented a computational and experimental pipeline to mine, screen, and characterize lncRNAs related to these two biological processes. Through analysis of publicly available Zika virus (ZIKV) infection-regulated Aedes aegypti transcriptomes, at least six lncRNAs were identified as being significantly upregulated in response to infection in various mosquito tissues. The roles of these ZIKV-regulated lncRNAs (designated Zinc1, Zinc2, Zinc3, Zinc9, Zinc10 and Zinc22), were further investigated by dsRNA-mediated silencing studies. Our results show that silencing of Zinc1, Zinc2, and Zinc22 renders mosquitoes significantly less permissive to ZIKV infection, while silencing of Zinc22 also reduces fecundity, indicating a potential role for Zinc22 in trade-offs between vector competence and reproduction. We also found that silencing of Zinc9 significantly increases fecundity but has no effect on ZIKV infection, suggesting that Zinc9 may be a negative regulator of oviposition. Our work demonstrates that some lncRNAs play host factor roles by facilitating viral infection in mosquitoes. We also show that lncRNAs can influence both mosquito reproduction and permissiveness to virus infection, two biological systems with important roles in mosquito vectorial capacity.
Collapse
Affiliation(s)
- Alexandros Belavilas-Trovas
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Laboratory of Molecular Biology and Genomics, Department of Biochemistry & Biotechnology, University of Thessaly, Larissa, Greece
| | - Spyros Tastsoglou
- DIANA-Lab, Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Mary Kefi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Mihra Tavadia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Kostas D Mathiopoulos
- Laboratory of Molecular Biology and Genomics, Department of Biochemistry & Biotechnology, University of Thessaly, Larissa, Greece
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
20
|
Chen TY, Bozic J, Mathias D, Smartt CT. Immune-related transcripts, microbiota and vector competence differ in dengue-2 virus-infected geographically distinct Aedes aegypti populations. Parasit Vectors 2023; 16:166. [PMID: 37208697 PMCID: PMC10199558 DOI: 10.1186/s13071-023-05784-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Vector competence in Aedes aegypti is influenced by various factors. Crucial new control methods can be developed by recognizing which factors affect virus and mosquito interactions. METHODS In the present study we used three geographically distinct Ae. aegypti populations and compared their susceptibility to infection by dengue virus serotype 2 (DENV-2). To identify any differences among the three mosquito populations, we evaluated expression levels of immune-related genes and assessed the presence of microbiota that might contribute to the uniqueness in their vector competence. RESULTS Based on the results from the DENV-2 competence study, we categorized the three geographically distinct Ae. aegypti populations into a refractory population (Vilas do Atlântico), a susceptible population (Vero) and a susceptible but low transmission population (California). The immune-related transcripts were highly expressed in the California population but not in the refractory population. However, the Rel-1 gene was upregulated in the Vilas do Atlântico population following ingestion of a non-infectious blood meal, suggesting the gene's involvement in non-viral responses, such as response to microbiota. Screening of the bacteria, fungi and flaviviruses revealed differences between populations, and any of these could be one of the factors that interfere with the vector competence. CONCLUSIONS The results reveal potential factors that might impact the virus and mosquito interaction, as well as influence the Ae. aegypti refractory phenotype.
Collapse
Affiliation(s)
- Tse-Yu Chen
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, University of Florida, Vero Beach, FL USA
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT USA
| | - Jovana Bozic
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, University of Florida, Vero Beach, FL USA
- Department of Entomology, The Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA USA
| | - Derrick Mathias
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, University of Florida, Vero Beach, FL USA
| | - Chelsea T. Smartt
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, University of Florida, Vero Beach, FL USA
| |
Collapse
|
21
|
Laureti M, Lee RX, Bennett A, Wilson LA, Sy VE, Kohl A, Dietrich I. Rift Valley Fever Virus Primes Immune Responses in Aedes aegypti Cells. Pathogens 2023; 12:563. [PMID: 37111448 PMCID: PMC10146816 DOI: 10.3390/pathogens12040563] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/25/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The ongoing global emergence of arthropod-borne (arbo) viruses has accelerated research into the interactions of these viruses with the immune systems of their vectors. Only limited information exists on how bunyaviruses, such as Rift Valley fever virus (RVFV), are sensed by mosquito immunity or escape detection. RVFV is a zoonotic phlebovirus (Bunyavirales; Phenuiviridae) of veterinary and human public health and economic importance. We have shown that the infection of mosquitoes with RVFV triggers the activation of RNA interference pathways, which moderately restrict viral replication. Here, we aimed to better understand the interactions between RVFV and other vector immune signaling pathways that might influence RVFV replication and transmission. For this, we used the immunocompetent Aedes aegypti Aag2 cell line as a model. We found that bacteria-induced immune responses restricted RVFV replication. However, virus infection alone did not alter the gene expression levels of immune effectors. Instead, it resulted in the marked enhancement of immune responses to subsequent bacterial stimulation. The gene expression levels of several mosquito immune pattern recognition receptors were altered by RVFV infection, which may contribute to this immune priming. Our findings imply that there is a complex interplay between RVFV and mosquito immunity that could be targeted in disease prevention strategies.
Collapse
Affiliation(s)
| | - Rui-Xue Lee
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Amelia Bennett
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- Department of Life Sciences, Faculty of Science, Claverton Down, University of Bath, Bath BA2 7AY, UK
| | - Lucas Aladar Wilson
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | | | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| | - Isabelle Dietrich
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK
- MRC-University of Glasgow Centre for Virus Research, 464 Bearsden Road, Glasgow G61 1QH, UK
| |
Collapse
|
22
|
Hussain M, Etebari K, Asgari S. Analysing inhibition of dengue virus in Wolbachia-infected mosquito cells following the removal of Wolbachia. Virology 2023; 581:48-55. [PMID: 36889142 DOI: 10.1016/j.virol.2023.02.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Wolbachia pipientis is known to block replication of positive sense RNA viruses. Previously, we created an Aedes aegypti Aag2 cell line (Aag2.wAlbB) transinfected with the wAlbB strain of Wolbachia and a matching tetracycline-cured Aag2.tet cell line. While dengue virus (DENV) was blocked in Aag2.wAlbB cells, we found significant inhibition of DENV in Aag2.tet cells. RNA-Seq analysis of the cells confirmed removal of Wolbachia and lack of expression of Wolbachia genes that could have been due to lateral gene transfer in Aag2.tet cells. However, we noticed a substantial increase in the abundance of phasi charoen-like virus (PCLV) in Aag2.tet cells. When RNAi was used to reduce the PCLV levels, DENV replication was significantly increased. Further, we found significant changes in the expression of antiviral and proviral genes in Aag2.tet cells. Overall, the results reveal an antagonistic interaction between DENV and PCLV and how PCLV-induced changes could contribute to DENV inhibition.
Collapse
Affiliation(s)
- Mazhar Hussain
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kayvan Etebari
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sassan Asgari
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
23
|
Alexander AJT, Salvemini M, Sreenu VB, Hughes J, Telleria EL, Ratinier M, Arnaud F, Volf P, Brennan B, Varjak M, Kohl A. Characterisation of the antiviral RNA interference response to Toscana virus in sand fly cells. PLoS Pathog 2023; 19:e1011283. [PMID: 36996243 PMCID: PMC10112792 DOI: 10.1371/journal.ppat.1011283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/18/2023] [Accepted: 03/09/2023] [Indexed: 04/01/2023] Open
Abstract
Toscana virus (TOSV) (Bunyavirales, Phenuiviridae, Phlebovirus, Toscana phlebovirus) and other related human pathogenic arboviruses are transmitted by phlebotomine sand flies. TOSV has been reported in nations bordering the Mediterranean Sea among other regions. Infection can result in febrile illness as well as meningitis and encephalitis. Understanding vector-arbovirus interactions is crucial to improving our knowledge of how arboviruses spread, and in this context, immune responses that control viral replication play a significant role. Extensive research has been conducted on mosquito vector immunity against arboviruses, with RNA interference (RNAi) and specifically the exogenous siRNA (exo-siRNA) pathway playing a critical role. However, the antiviral immunity of phlebotomine sand flies is less well understood. Here we were able to show that the exo-siRNA pathway is active in a Phlebotomus papatasi-derived cell line. Following TOSV infection, distinctive 21 nucleotide virus-derived small interfering RNAs (vsiRNAs) were detected. We also identified the exo-siRNA effector Ago2 in this cell line, and silencing its expression rendered the exo-siRNA pathway largely inactive. Thus, our data show that this pathway is active as an antiviral response against a sand fly transmitted bunyavirus, TOSV.
Collapse
Affiliation(s)
| | - Marco Salvemini
- Department of Biology, University of Naples Federico II, Italy
| | | | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Erich L. Telleria
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Maxime Ratinier
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon1, EPHE, PSL Research University, Lyon, France
| | - Frédérick Arnaud
- IVPC UMR754, INRAE, Univ Lyon, Université Claude Bernard Lyon1, EPHE, PSL Research University, Lyon, France
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Benjamin Brennan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Margus Varjak
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
24
|
Insect-Specific Chimeric Viruses Potentiated Antiviral Responses and Inhibited Pathogenic Alphavirus Growth in Mosquito Cells. Microbiol Spectr 2023; 11:e0361322. [PMID: 36511715 PMCID: PMC9927327 DOI: 10.1128/spectrum.03613-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Most alphaviruses are transmitted by mosquito vectors and infect a wide range of vertebrate hosts, with a few exceptions. Eilat virus (EILV) in this genus is characterized by a host range restricted to mosquitoes. Its chimeric viruses have been developed as safe and effective vaccine candidates and diagnostic tools. Here, we investigated the interactions between these insect-specific viruses (ISVs) and mosquito cells, unveiling their potential roles in determining vector competence and arbovirus transmission. By RNA sequencing, we found that these ISVs profoundly modified host cell gene expression profiles. Two EILV-based chimeras, consisting of EILV's nonstructural genes and the structural genes of Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV), namely, EILV/CHIKV (E/C) and EILV/VEEV (E/V), induced more intensive transcriptome regulation than parental EILV and activated different antiviral mechanisms in host cells. We demonstrated that E/C robustly promoted antimicrobial peptide production and E/V strongly upregulated the RNA interference pathway components. This also highlighted the intrinsic divergences between CHIKV and VEEV, representatives of the Old World and New World alphaviruses. In contrast, EILV triggered a limited antiviral response. We further showed that initial chimera infections efficiently inhibited subsequent pathogenic alphavirus replication, especially in the case of E/V infection, which almost prevented VEEV and Sindbis virus (SINV) superinfections. Altogether our study provided valuable information on developing ISVs as biological control agents. IMPORTANCE Mosquito-borne alphaviruses can cause emerging and reemerging infectious diseases, posing a considerable threat to human and animal health worldwide. However, no specific antivirals or commercial vaccines are currently available. Therefore, it is vital to develop biological control measures to contain virus transmission. Insect-specific EILV and its chimeras are supposed to induce superinfection exclusion owing to the close phylogenetical relationship with pathogenic alphaviruses. These viruses might also, like bacterial symbionts, modulate mosquito hosts' vector competence for arboviruses. However, little is known about the responses of mosquitoes or mosquito cells to ISV infections. Here, we found that EILV barely elicited antiviral defenses in host cells, while its chimeras, namely, E/C and E/V, potentiated the responses via different mechanisms. Furthermore, we showed that initial chimera infections could largely inhibit subsequent pathogenic alphavirus infections. Taken together, our study proposed insect-specific chimeras as a promising candidate for developing biological control measures against pathogenic alphaviruses.
Collapse
|
25
|
García-Longoria L, Ahrén D, Berthomieu A, Kalbskopf V, Rivero A, Hellgren O. Immune gene expression in the mosquito vector Culex quinquefasciatus during an avian malaria infection. Mol Ecol 2023; 32:904-919. [PMID: 36448733 PMCID: PMC10108303 DOI: 10.1111/mec.16799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Plasmodium relictum is the most widespread avian malaria parasite in the world. It is listed as one of the 100 most dangerous invasive species, having been responsible for the extinction of several endemic bird species, and the near-demise of several others. Here we present the first transcriptomic study focused on the effect of P. relictum on the immune system of its vector (the mosquito Culex quinquefasciatus) at different times post-infection. We show that over 50% of immune genes identified as being part of the Toll pathway and 30%-40% of the immune genes identified within the Imd pathway are overexpressed during the critical period spanning the parasite's oocyst and sporozoite formation (8-12 days), revealing the crucial role played by both these pathways in this natural mosquito-Plasmodium combination. Comparison of infected mosquitoes with their uninfected counterparts also revealed some unexpected immune RNA expression patterns earlier and later in the infection: significant differences in expression of several immune effectors were observed as early as 30 min after ingestion of the infected blood meal. In addition, in the later stages of the infection (towards the end of the mosquito lifespan), we observed an unexpected increase in immune investment in uninfected, but not in infected, mosquitoes. In conclusion, our work extends the comparative transcriptomic analyses of malaria-infected mosquitoes beyond human and rodent parasites and provides insights into the degree of conservation of immune pathways and into the selective pressures exerted by Plasmodium parasites on their vectors.
Collapse
Affiliation(s)
- Luz García-Longoria
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, Badajoz, Spain
| | - Dag Ahrén
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | | | - Victor Kalbskopf
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Ana Rivero
- MIVEGEC (CNRS, Université de Montpellier, IRD), Montpellier, France
| | - Olof Hellgren
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| |
Collapse
|
26
|
Morejon B, Michel K. A zone-of-inhibition assay to screen for humoral antimicrobial activity in mosquito hemolymph. Front Cell Infect Microbiol 2023; 13:891577. [PMID: 36779191 PMCID: PMC9908765 DOI: 10.3389/fcimb.2023.891577] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
In insects, antibacterial immunity largely depends on the activation of downstream signaling and effector responses, leading to the synthesis and secretion of soluble effector molecules, such as antimicrobial peptides (AMPs). AMPs are acute infection response peptides secreted into the hemolymph upon bacterial stimulation. The transcription of innate immunity genes encoding for AMPs is highly dependent on several signaling cascade pathways, such as the Toll pathway. In the African malaria mosquito, Anopheles gambiae, AMPs hold a special interest as their upregulation have been shown to limit the growth of malaria parasites, bacteria, and fungi. Most of the current knowledge on the regulation of insect AMPs in microbial infection have been obtained from Drosophila. However, largely due to the lack of convenient assays, the regulation of antimicrobial activity in mosquito hemolymph is still not completely understood. In this study, we report a zone of inhibition assay to identify the contribution of AMPs and components of the Toll pathway to the antimicrobial activity of A. gambiae hemolymph. As a proof of principle, we demonstrate that Micrococcus luteus challenge induces antimicrobial activity in the adult female mosquito hemolymph, which is largely dependent on defensin 1. Moreover, by using RNAi to silence Cactus, REL1, and MyD88, we showed that Cactus kd induces antimicrobial activity in the mosquito hemolymph, whereas the antimicrobial activity in REL1 kd and MyD88 kd is reduced after challenge. Finally, while injection itself is not sufficient to induce antimicrobial activity, our results show that it primes the response to bacterial challenge. Our study provides information that increases our knowledge of the regulation of antimicrobial activity in response to microbial infections in mosquitoes. Furthermore, this assay represents an ex vivo medium throughput assay that can be used to determine the upstream regulatory elements of antimicrobial activity in A. gambiae hemolymph.
Collapse
Affiliation(s)
- Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | | |
Collapse
|
27
|
Bui M, Dalla Benetta E, Dong Y, Zhao Y, Yang T, Li M, Antoshechkin IA, Buchman A, Bottino-Rojas V, James AA, Perry MW, Dimopoulos G, Akbari OS. CRISPR mediated transactivation in the human disease vector Aedes aegypti. PLoS Pathog 2023; 19:e1010842. [PMID: 36656895 PMCID: PMC9888728 DOI: 10.1371/journal.ppat.1010842] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/31/2023] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
As a major insect vector of multiple arboviruses, Aedes aegypti poses a significant global health and economic burden. A number of genetic engineering tools have been exploited to understand its biology with the goal of reducing its impact. For example, current tools have focused on knocking-down RNA transcripts, inducing loss-of-function mutations, or expressing exogenous DNA. However, methods for transactivating endogenous genes have not been developed. To fill this void, here we developed a CRISPR activation (CRISPRa) system in Ae. aegypti to transactivate target gene expression. Gene expression is activated through pairing a catalytically-inactive ('dead') Cas9 (dCas9) with a highly-active tripartite activator, VP64-p65-Rta (VPR) and synthetic guide RNA (sgRNA) complementary to a user defined target-gene promoter region. As a proof of concept, we demonstrate that engineered Ae. aegypti mosquitoes harboring a binary CRISPRa system can be used to effectively overexpress two developmental genes, even-skipped (eve) and hedgehog (hh), resulting in observable morphological phenotypes. We also used this system to overexpress the positive transcriptional regulator of the Toll immune pathway known as AaRel1, which resulted in a significant suppression of dengue virus serotype 2 (DENV2) titers in the mosquito. This system provides a versatile tool for research pathways not previously possible in Ae. aegypti, such as programmed overexpression of endogenous genes, and may aid in gene characterization studies and the development of innovative vector control tools.
Collapse
Affiliation(s)
- Michelle Bui
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Elena Dalla Benetta
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yunchong Zhao
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Ting Yang
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Ming Li
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Igor A. Antoshechkin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Anna Buchman
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Vanessa Bottino-Rojas
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Anthony A. James
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, California, United States of America
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Michael W. Perry
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Omar S. Akbari
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
28
|
Carvalho KS, Rezende TMT, Romão TP, Rezende AM, Chiñas M, Guedes DRD, Paiva-Cavalcanti M, Silva-Filha MHNL. Aedes aegypti Strain Subjected to Long-Term Exposure to Bacillus thuringiensis svar. israelensis Larvicides Displays an Altered Transcriptional Response to Zika Virus Infection. Viruses 2022; 15:72. [PMID: 36680112 PMCID: PMC9866606 DOI: 10.3390/v15010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Bacillus thuringiensis svar. israelensis (Bti) larvicides are effective in controlling Aedes aegypti; however, the effects of long-term exposure need to be properly evaluated. We established an Ae. aegypti strain that has been treated with Bti for 30 generations (RecBti) and is still susceptible to Bti, but females exhibited increased susceptibility to Zika virus (ZIKV). This study compared the RecBti strain to a reference strain regarding: first, the relative transcription of selected immune genes in ZIKV-challenged females (F30) with increased susceptibility detected in a previous study; then, the whole transcriptomic profile using unchallenged females (F35). Among the genes compared by RT-qPCR in the ZIKV-infected and uninfected females from RecBti (F30) and the reference strain, hop, domeless, relish 1, defensin A, cecropin D, and gambicin showed a trend of repression in RecBti infected females. The transcriptome of RecBti (F35) unchallenged females, compared with a reference strain by RNA-seq, showed a similar profile and only 59 differentially expressed genes were found among 9202 genes analyzed. Our dataset showed that the long-term Bti exposure of the RecBti strain was associated with an alteration of the expression of genes potentially involved in the response to ZIKV infection in challenged females, which is an important feature found under this condition.
Collapse
Affiliation(s)
- Karine S. Carvalho
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, Brazil
| | | | - Tatiany P. Romão
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, Brazil
| | - Antônio M. Rezende
- Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife 50670-420, Brazil
| | - Marcos Chiñas
- Center for Genomic Sciences, National Autonomous University of Mexico, Cuernavaca 62210, Mexico
| | | | | | | |
Collapse
|
29
|
Cardoso-Jaime V, Tikhe CV, Dong S, Dimopoulos G. The Role of Mosquito Hemocytes in Viral Infections. Viruses 2022; 14:v14102088. [PMID: 36298644 PMCID: PMC9608948 DOI: 10.3390/v14102088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Insect hemocytes are the only immune cells that can mount a humoral and cellular immune response. Despite the critical involvement of hemocytes in immune responses against bacteria, fungi, and parasites in mosquitoes, our understanding of their antiviral potential is still limited. It has been shown that hemocytes express humoral factors such as TEP1, PPO, and certain antimicrobial peptides that are known to restrict viral infections. Insect hemocytes also harbor the major immune pathways, such as JAK/STAT, TOLL, IMD, and RNAi, which are critical for the control of viral infection. Recent research has indicated a role for hemocytes in the regulation of viral infection through RNA interference and autophagy; however, the specific mechanism by which this regulation occurs remains uncharacterized. Conversely, some studies have suggested that hemocytes act as agonists of arboviral infection because they lack basal lamina and circulate throughout the whole mosquito, likely facilitating viral dissemination to other tissues such as salivary glands. In addition, hemocytes produce arbovirus agonist factors such as lectins, which enhance viral infection. Here, we summarize our current understanding of hemocytes’ involvement in viral infections.
Collapse
|
30
|
Tikhe CV, Cardoso-Jaime V, Dong S, Rutkowski N, Dimopoulos G. Trypsin-like Inhibitor Domain (TIL)-Harboring Protein Is Essential for Aedes aegypti Reproduction. Int J Mol Sci 2022; 23:ijms23147736. [PMID: 35887084 PMCID: PMC9319116 DOI: 10.3390/ijms23147736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023] Open
Abstract
Cysteine-rich trypsin inhibitor-like domain (TIL)-harboring proteins are broadly distributed in nature but remain understudied in vector mosquitoes. Here we have explored the biology of a TIL domain-containing protein of the arbovirus vector Aedes aegypti, cysteine-rich venom protein 379 (CRVP379). CRVP379 was previously shown to be essential for dengue virus infection in Ae. aegypti mosquitoes. Gene expression analysis showed CRVP379 to be highly expressed in pupal stages, male testes, and female ovaries. CRVP379 expression is also increased in the ovaries at 48 h post-blood feeding. We used CRISPR-Cas9 genome editing to generate two mutant lines of CRVP379 with mutations inside or outside the TIL domain. Female mosquitoes from both mutant lines showed severe defects in their reproductive capability; mutant females also showed differences in their follicular cell morphology. However, the CRVP379 line with a mutation outside the TIL domain did not affect male reproductive performance, suggesting that some CRVP379 residues may have sexually dimorphic functions. In contrast to previous reports, we did not observe a noticeable difference in dengue virus infection between the wild-type and any of the mutant lines. The importance of CRVP379 in Ae. aegypti reproductive biology makes it an interesting candidate for the development of Ae. aegypti population control methods.
Collapse
Affiliation(s)
- Chinmay Vijay Tikhe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.V.T.); (V.C.-J.); (S.D.); (N.R.)
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.V.T.); (V.C.-J.); (S.D.); (N.R.)
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.V.T.); (V.C.-J.); (S.D.); (N.R.)
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Natalie Rutkowski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.V.T.); (V.C.-J.); (S.D.); (N.R.)
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (C.V.T.); (V.C.-J.); (S.D.); (N.R.)
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Correspondence:
| |
Collapse
|
31
|
Amarante ADM, da Silva ICDA, Carneiro VC, Vicentino ARR, Pinto MDA, Higa LM, Moharana KC, Talyuli OAC, Venancio TM, de Oliveira PL, Fantappié MR. Zika virus infection drives epigenetic modulation of immunity by the histone acetyltransferase CBP of Aedes aegypti. PLoS Negl Trop Dis 2022; 16:e0010559. [PMID: 35759510 PMCID: PMC9269902 DOI: 10.1371/journal.pntd.0010559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/08/2022] [Accepted: 06/03/2022] [Indexed: 11/18/2022] Open
Abstract
Epigenetic mechanisms are responsible for a wide range of biological phenomena in insects, controlling embryonic development, growth, aging and nutrition. Despite this, the role of epigenetics in shaping insect-pathogen interactions has received little attention. Gene expression in eukaryotes is regulated by histone acetylation/deacetylation, an epigenetic process mediated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). In this study, we explored the role of the Aedes aegypti histone acetyltransferase CBP (AaCBP) after infection with Zika virus (ZIKV), focusing on the two main immune tissues, the midgut and fat body. We showed that the expression and activity of AaCBP could be positively modulated by blood meal and ZIKV infection. Nevertheless, Zika-infected mosquitoes that were silenced for AaCBP revealed a significant reduction in the acetylation of H3K27 (CBP target marker), followed by downmodulation of the expression of immune genes, higher titers of ZIKV and lower survival rates. Importantly, in Zika-infected mosquitoes that were treated with sodium butyrate, a histone deacetylase inhibitor, their capacity to fight virus infection was rescued. Our data point to a direct correlation among histone hyperacetylation by AaCBP, upregulation of antimicrobial peptide genes and increased survival of Zika-infected-A. aegypti.
Collapse
Affiliation(s)
- Anderson de Mendonça Amarante
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Isabel Caetano de Abreu da Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Vitor Coutinho Carneiro
- Division of Epigenetics, German Cancer Research Center, Im Neuenheimer Feld, Heidelberg, Germany
| | - Amanda Roberta Revoredo Vicentino
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcia de Amorim Pinto
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Luiza Mendonça Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Kanhu Charan Moharana
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brasil
| | - Octavio A. C. Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Thiago Motta Venancio
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Brasil
| | - Pedro Lagerblad de Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcelo Rosado Fantappié
- Instituto de Bioquímica Médica Leopoldo de Meis, Programa de Biologia Molecular e Biotecnologia, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
32
|
Fiorillo C, Yen PS, Colantoni A, Mariconti M, Azevedo N, Lombardo F, Failloux AB, Arcà B. MicroRNAs and other small RNAs in Aedes aegypti saliva and salivary glands following chikungunya virus infection. Sci Rep 2022; 12:9536. [PMID: 35681077 PMCID: PMC9184468 DOI: 10.1038/s41598-022-13780-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
Mosquito saliva facilitates blood feeding through the anti-haemostatic, anti-inflammatory and immunomodulatory properties of its proteins. However, the potential contribution of non-coding RNAs to host manipulation is still poorly understood. We analysed small RNAs from Aedes aegypti saliva and salivary glands and show here that chikungunya virus-infection triggers both the siRNA and piRNA antiviral pathways with limited effects on miRNA expression profiles. Saliva appears enriched in specific miRNA subsets and its miRNA content is well conserved among mosquitoes and ticks, clearly pointing to a non-random sorting and occurrence. Finally, we provide evidence that miRNAs from Ae. aegypti saliva may target human immune and inflammatory pathways, as indicated by prediction analysis and searching for experimentally validated targets of identical human miRNAs. Overall, we believe these observations convincingly support a scenario where both proteins and miRNAs from mosquito saliva are injected into vertebrates during blood feeding and contribute to the complex vector-host-pathogen interactions.
Collapse
Affiliation(s)
- Carmine Fiorillo
- Department of Public Health and Infectious Diseases - Division of Parasitology, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Pei-Shi Yen
- Arboviruses and Insect Vectors Unit, Institute Pasteur, 25 rue Dr. Roux, 75724, Paris Cedex 15, France
| | - Alessio Colantoni
- Department of Biology and Biotechnology, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Marina Mariconti
- Arboviruses and Insect Vectors Unit, Institute Pasteur, 25 rue Dr. Roux, 75724, Paris Cedex 15, France
| | - Nayara Azevedo
- Genomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases - Division of Parasitology, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Unit, Institute Pasteur, 25 rue Dr. Roux, 75724, Paris Cedex 15, France
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases - Division of Parasitology, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
33
|
Zhang XZ, Li FH, Wang XJ. Regulation of Tripartite Motif-Containing Proteins on Immune Response and Viral Evasion. Front Microbiol 2021; 12:794882. [PMID: 34925304 PMCID: PMC8671828 DOI: 10.3389/fmicb.2021.794882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/08/2021] [Indexed: 12/21/2022] Open
Abstract
Tripartite motif-containing proteins (TRIMs), exhibiting ubiquitin E3 ligase activity, are involved in regulation of not only autophagy and apoptosis but also pyrotosis and antiviral immune responses of host cells. TRIMs play important roles in modulating signaling pathways of antiviral immune responses via type I interferon, NF-κB, Janus kinase/signal transducer and activator of transcription (JAK/STAT), and Nrf2. However, viruses are able to antagonize TRIM activity or evenly utilize TRIMs for viral replication. This communication presents the current understanding of TRIMs exploited by viruses to evade host immune response.
Collapse
Affiliation(s)
- Xiu-Zhong Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Fu-Huang Li
- Beijing General Station of Animal Husbandry Service (South Section), Beijing, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
34
|
Enoxacin shows a broad-spectrum antiviral activity against diverse viruses by enhancing antiviral RNAi in insects. J Virol 2021; 96:e0177821. [PMID: 34908449 DOI: 10.1128/jvi.01778-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RNA interference (RNAi) functions as the major host antiviral defense in insects, while less is understood about how to utilize antiviral RNAi in controlling viral infection in insects. Enoxacin belongs to the family of synthetic antibacterial compounds based on a fluoroquinolone skeleton that has been previously found to enhance RNAi in mammalian cells. In this study, we showed that enoxacin efficiently inhibited viral replication of Drosophila C virus (DCV) and Cricket paralysis virus (CrPV) in cultured Drosophila cells. Enoxacin promoted the loading of Dicer-2-processed virus-derived siRNA into the RNA-induced silencing complex, thereby enhancing antiviral RNAi response in infected cells. Moreover, enoxacin treatment elicited an RNAi-dependent in vivo protective efficacy against DCV or CrPV challenge in adult fruit flies. In addition, enoxacin also inhibited replication of flaviviruses, including Dengue virus and Zika virus, in Aedes mosquito cells in an RNAi-dependent manner. Together, our findings demonstrated that enoxacin can enhance RNAi in insects, and enhancing RNAi by enoxacin is an effective antiviral strategy against diverse viruses in insects, which may be exploited as a broad-spectrum antiviral agent to control vector transmission of arboviruses or viral diseases in insect farming. Importance RNAi has been widely recognized as one of the most broadly acting and robust antiviral mechanism in insects. However, the application of antiviral RNAi in controlling viral infections in insects is less understood. Enoxacin is a fluoroquinolone compound that has been previously found to enhance RNAi in mammalian cells, while its RNAi-enhancing activity has not been assessed in insects. Herein, we showed that enoxacin treatment inhibited viral replication of DCV and CrPV in Drosophila cells and in adult fruit flies. Enoxacin promoted the loading of Dicer-generated virus-derived siRNA into Ago2-incorporated RNA-induced silencing complex, and in turn strengthened the antiviral RNAi response in the infected cells. Moreover, enoxacin also displayed effective RNAi-dependent antiviral effects against flaviviruses, such as Dengue virus and Zika virus, in mosquito cells. This study is the first to demonstrate that enhancing RNAi by enoxacin elicits potent antiviral efficacies against diverse viruses in insects.
Collapse
|
35
|
Xu Y, Zhong Z, Ren Y, Ma L, Ye Z, Gao C, Wang J, Li Y. Antiviral RNA interference in disease vector (Asian longhorned) ticks. PLoS Pathog 2021; 17:e1010119. [PMID: 34860862 PMCID: PMC8673602 DOI: 10.1371/journal.ppat.1010119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 12/15/2021] [Accepted: 11/16/2021] [Indexed: 11/18/2022] Open
Abstract
Disease vectors such as mosquitoes and ticks play a major role in the emergence and re-emergence of human and animal viral pathogens. Compared to mosquitoes, however, much less is known about the antiviral responses of ticks. Here we showed that Asian longhorned ticks (Haemaphysalis longicornis) produced predominantly 22-nucleotide virus-derived siRNAs (vsiRNAs) in response to severe fever with thrombocytopenia syndrome virus (SFTSV, an emerging tick-borne virus), Nodamura virus (NoV), or Sindbis virus (SINV) acquired by blood feeding. Notably, experimental acquisition of NoV and SINV by intrathoracic injection also initiated viral replication and triggered the production of vsiRNAs in H. longicornis. We demonstrated that a mutant NoV deficient in expressing its viral suppressor of RNAi (VSR) replicated to significantly lower levels than wildtype NoV in H. longicornis, but accumulated to higher levels after knockdown of the tick Dicer2-like protein identified by phylogeny comparison. Moreover, the expression of a panel of known animal VSRs in cis from the genome of SINV drastically enhanced the accumulation of the recombinant viruses. This study establishes a novel model for virus-vector-mouse experiments with longhorned ticks and provides the first in vivo evidence for an antiviral function of the RNAi response in ticks. Interestingly, comparing the accumulation levels of SINV recombinants expressing green fluorescent protein or SFTSV proteins identified the viral non-structural protein as a putative VSR. Elucidating the function of ticks’ antiviral RNAi pathway in vivo is critical to understand the virus-host interaction and the control of tick-borne viral pathogens. Tick-borne diseases (TBDs) are the most common illnesses transmitted by ticks, and the annual number of reported TBD cases continues to increase. The Asian longhorned tick, a vector associated with at least 30 human pathogens, is native to eastern Asia and recently reached the USA as an emerging disease threat. Newly identified tick-transmitted pathogens continue to be reported, raising concerns about how TBDs occur. Interestingly, tick can harbor pathogens without being affected themselves. For viral infections, ticks have their own immune systems that protect them from infection. Meanwhile, tick-borne viruses have evolved to avoid these defenses as they establish themselves within the vector. Here, we show in detail that infecting longhorned ticks with distinct arthropod-borne RNA viruses through two approaches natural blood feeding and injection, all induce the production of vsiRNAs. Dicer2-like homolog plays a role in regulating antiviral RNAi responses as knocking down of this gene enhanced viral replication. Furthermore, we demonstrate that tick antiviral RNAi responses are inhibited through expression heterologous VSR proteins in recombinant SINV. We identify both the virus and tick factors are critical components to understanding TBDs. Importantly, our study introduces a novel, in vivo virus-vector-mouse model system for exploring TBDs in the future.
Collapse
Affiliation(s)
- Yan Xu
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhengwei Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanxin Ren
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Liting Ma
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhi Ye
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Chuang Gao
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- * E-mail: (JW); (YL)
| | - Yang Li
- CAS Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- * E-mail: (JW); (YL)
| |
Collapse
|
36
|
Angleró-Rodríguez YI, Tikhe CV, Kang S, Dimopoulos G. Aedes aegypti Toll pathway is induced through dsRNA sensing in endosomes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104138. [PMID: 34022257 DOI: 10.1016/j.dci.2021.104138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 06/12/2023]
Abstract
Mosquito anti-pathogen immune responses, including those controlling infection with arboviruses are regulated by multiple signal transduction pathways. While the Toll pathway is critical in the defense against arboviruses such as dengue and Zika viruses, the factors and mechanisms involved in virus recognition leading to the activation of the Toll pathway are not fully understood. In this study we evaluated the role of virus-produced double-stranded RNA (dsRNA) intermediates in mosquito immune activation by utilizing the synthetic dsRNA analog polyinosinic-polycytidylic acid (poly I:C). Poly I:C treatment of Aedes aegypti mosquitoes and Aag2 cells reduced DENV infection. Transcriptomic analyses of Aag2 cell responses to poly I:C indicated putative activation of the Toll pathway. We found that poly I:C is translocated to the endosomal compartment of Aag2 cells, and that the A. aegypti Toll 6 receptor is a putative dsRNA recognition receptor. This study elucidates the role of dsRNAs in the immune activation of non-RNAi pathways in mosquitoes.
Collapse
Affiliation(s)
| | - Chinmay V Tikhe
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, United States
| | - Seokyoung Kang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, United States
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, United States.
| |
Collapse
|
37
|
Chen TY, Lee Y, Wang X, Mathias D, Caragata EP, Smartt CT. Profiling Transcriptional Response of Dengue-2 Virus Infection in Midgut Tissue of Aedes aegypti. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.708817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Understanding the mosquito antiviral response could reveal target pathways or genes of interest that could form the basis of new disease control applications. However, there is a paucity of data in the current literature in understanding antiviral response during the replication period. To illuminate the gene expression patterns in the replication stage, we collected gene expression data at 2.5 days after Dengue-2 virus (DENV-2) infection. We sequenced the whole transcriptome of the midgut tissue and compared gene expression levels between the control and virus-infected group. We identified 31 differentially expressed genes. Based on their function, we identified that those genes fell into two major functional categories - (1) nucleic acid/protein process and (2) immunity/oxidative stress response. Our study has identified candidate genes that can be followed up for gene overexpression/inhibition experiments to examine if the perturbed gene interaction may impact the mosquito’s immune response against DENV. This is an important step to understanding how mosquitoes eliminate the virus and provides an important foundation for further research in developing novel dengue control strategies.
Collapse
|
38
|
Gabrieli P, Caccia S, Varotto-Boccazzi I, Arnoldi I, Barbieri G, Comandatore F, Epis S. Mosquito Trilogy: Microbiota, Immunity and Pathogens, and Their Implications for the Control of Disease Transmission. Front Microbiol 2021; 12:630438. [PMID: 33889137 PMCID: PMC8056039 DOI: 10.3389/fmicb.2021.630438] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
In mosquitoes, the interaction between the gut microbiota, the immune system, and the pathogens that these insects transmit to humans and animals is regarded as a key component toward the development of control strategies, aimed at reducing the burden of severe diseases, such as malaria and dengue fever. Indeed, different microorganisms from the mosquito microbiota have been investigated for their ability to affect important traits of the biology of the host insect, related with its survival, development and reproduction. Furthermore, some microorganisms have been shown to modulate the immune response of mosquito females, significantly shaping their vector competence. Here, we will review current knowledge in this field, focusing on i) the complex interaction between the intestinal microbiota and mosquito females defenses, both in the gut and at humoral level; ii) how knowledge on these issues contributes to the development of novel and targeted strategies for the control of mosquito-borne diseases such as the use of paratransgenesis or taking advantage of the relationship between Wolbachia and mosquito hosts. We conclude by providing a brief overview of available knowledge on microbiota-immune system interplay in major insect vectors.
Collapse
Affiliation(s)
- Paolo Gabrieli
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Silvia Caccia
- Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Irene Arnoldi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Francesco Comandatore
- "L. Sacco" Department of Biomedical and Clinical Sciences, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|