1
|
Estévez-Lao TY, Martin LE, Hillyer JF. Activation of the immune deficiency pathway (IMD) reduces the mosquito heart rate via a nitric oxide-based mechanism. JOURNAL OF INSECT PHYSIOLOGY 2025; 161:104738. [PMID: 39647603 DOI: 10.1016/j.jinsphys.2024.104738] [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: 09/10/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
The immune deficiency pathway (IMD) is an important component of the antibacterial, antimalarial and antiviral response in mosquitoes. The IMD pathway also drives the infection induced migration of hemocytes to the heart. During an infection, periostial hemocytes kill pathogens in areas of high hemolymph flow and produce nitric oxide that reduces the heart rate. Here, we investigated the consequences of repressing the IMD pathway by silencing the transcription factor, rel2, or activating the pathway by silencing the negative regulator, caspar, in Anopheles gambiae. In uninfected mosquitoes, repression of the IMD pathway does not affect the circulatory system. However, activating the IMD pathway decreases the heart rate, and this correlates with increased transcription and activity of nitric oxide synthase (NOS), but not increased transcription of the lysozymes, LysC1 or LysC2. In infected mosquitoes, however, activation of the IMD pathway does not affect the heart rate but repression of the pathway decreases the heart rate. This latter phenotype correlates with increased transcription and activity of nitric oxide synthase, which is likely due to an increase in infection intensity. In conclusion, we demonstrate that a major immune signaling pathway that regulates periostial hemocyte aggregation, the IMD pathway, reduces the heart rate via a nitric oxide-based mechanism.
Collapse
Affiliation(s)
- Tania Y Estévez-Lao
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Lindsay E Martin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Julián F Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Rhodes VL, Waterhouse RM, Michel K. The molecular toll pathway repertoire in anopheline mosquitoes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105287. [PMID: 39522894 PMCID: PMC11717629 DOI: 10.1016/j.dci.2024.105287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Innate immunity in mosquitoes has received much attention due to its potential impact on vector competence for vector-borne disease pathogens, including malaria parasites. The nuclear factor (NF)-κB-dependent Toll pathway is a major regulator of innate immunity in insects. In mosquitoes, this pathway controls transcription of the majority of the known canonical humoral immune effectors, mediates anti-bacterial, anti-fungal and anti-viral immune responses, and contributes to malaria parasite killing. However, besides initial gene annotation of putative Toll pathway members and genetic analysis of the contribution of few key components to immunity, the molecular make-up and function of the Toll pathway in mosquitoes is largely unexplored. To facilitate functional analyses of the Toll pathway in mosquitoes, we report here manually annotated and refined gene models of Toll-like receptors and all putative components of the intracellular signal transduction cascade across 19 anopheline genomes, and in two culicine genomes. In addition, based on phylogenetic analyses, we identified differing levels of evolutionary constraint across the intracellular Toll pathway members, and identified a recent radiation of TOLL1/5 within the Anopheles gambiae complex. Together, this study provides insight into the evolution of TLRs and the putative members of the intracellular signal transduction cascade within the genus Anopheles.
Collapse
Affiliation(s)
- Victoria L Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA; Biology Department, Missouri Southern University, Joplin, MO 64801, USA
| | | | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
3
|
Shirozu T, Regilme MAF, Ote M, Sasaki M, Soga A, Bochimoto H, Kawabata H, Umemiya-Shirafuji R, Kanuka H, Fukumoto S. Wolbachia infection in Aedes aegypti does not affect its vectorial capacity for Dirofilaria immitis. Sci Rep 2024; 14:22528. [PMID: 39341970 PMCID: PMC11439018 DOI: 10.1038/s41598-024-73421-9] [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: 05/10/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Mosquito-borne diseases such as dengue and filariasis are a growing public health concern in endemic countries. Biological approaches, such as the trans-infection of Wolbachia pipientis in mosquitoes, are an alternative vector control strategy, especially for arthropod-borne viruses such as dengue. In the present study, the effect of Wolbachia (wMel strain) on the vectorial capacity of Aedes aegypti for Dirofilaria immitis was studied. Our results showed that Wolbachia does not affect the phenotype of mosquito survival or the prevalence, number, and molting rate of third-stage larvae in both susceptible and resistant strains of Ae. aegypti. RNA-seq analysis of Malpighian tubules at 2 days post-infection with D. immitis showed the differentially expressed genes (DEGs) with and without wMel infection. No characteristic immune-related gene expression patterns were observed among the DEGs. No significant change in the amount of Wolbachia was observed in the Ae. aegypti after D. immitis infection. Our results suggest that infection of D. immitis in Ae. aegypti populations will not interfere with Wolbachia-based vector control strategies in dengue-endemic areas where cases of D. immitis are present. This study demonstrated the veterinary medical validity of a dengue control program using Wolbachia.
Collapse
Affiliation(s)
- Takahiro Shirozu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Maria Angenica F Regilme
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Manabu Ote
- Department of Tropical Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
- Center for Medical Entomology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Mizuki Sasaki
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Akira Soga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hiroki Bochimoto
- Department of Cell Physiology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hidenobu Kawabata
- Health Care Administration Center, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Rika Umemiya-Shirafuji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hirotaka Kanuka
- Department of Tropical Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
- Center for Medical Entomology, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Shinya Fukumoto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
4
|
Hixson B, Chen R, Buchon N. Innate immunity in Aedes mosquitoes: from pathogen resistance to shaping the microbiota. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230063. [PMID: 38497256 PMCID: PMC10945403 DOI: 10.1098/rstb.2023.0063] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/12/2023] [Indexed: 03/19/2024] Open
Abstract
Discussions of host-microbe interactions in mosquito vectors are frequently dominated by a focus on the human pathogens they transmit (e.g. Plasmodium parasites and arboviruses). Underlying the interactions between a vector and its transmissible pathogens, however, is the physiology of an insect living and interacting with a world of bacteria and fungi including commensals, mutualists and primary and opportunistic pathogens. Here we review what is known about the bacteria and fungi associated with mosquitoes, with an emphasis on the members of the Aedes genus. We explore the reciprocal effects of microbe on mosquito, and mosquito on microbe. We analyse the roles of bacterial and fungal symbionts in mosquito development, their effects on vector competence, and their potential uses as biocontrol agents and vectors for paratransgenesis. We explore the compartments of the mosquito gut, uncovering the regionalization of immune effectors and modulators, which create the zones of resistance and immune tolerance with which the mosquito host controls and corrals its microbial symbionts. We examine the anatomical patterning of basally expressed antimicrobial peptides. Finally, we review the relationships between inducible antimicrobial peptides and canonical immune signalling pathways, comparing and contrasting current knowledge on each pathway in mosquitoes to the model insect Drosophila melanogaster. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Bretta Hixson
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| | - Robin Chen
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| |
Collapse
|
5
|
Hixson B, Huot L, Morejon B, Yang X, Nagy P, Michel K, Buchon N. The transcriptional response in mosquitoes distinguishes between fungi and bacteria but not Gram types. BMC Genomics 2024; 25:353. [PMID: 38594632 PMCID: PMC11003161 DOI: 10.1186/s12864-024-10153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/22/2024] [Indexed: 04/11/2024] Open
Abstract
Mosquitoes are prolific vectors of human pathogens, therefore a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster, is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae (s.l.) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti, however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.
Collapse
Affiliation(s)
- Bretta Hixson
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Louise Huot
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Xiaowei Yang
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
- Current address: State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute for Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Peter Nagy
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Nicolas Buchon
- Cornell Institute of Host-Microbe Interactions and Disease, Department of Entomology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
6
|
Peng J, Zhang M, Wang G, Zhang D, Zheng X, Li Y. Biased virus transmission following sequential coinfection of Aedes aegypti with dengue and Zika viruses. PLoS Negl Trop Dis 2024; 18:e0012053. [PMID: 38557981 PMCID: PMC10984552 DOI: 10.1371/journal.pntd.0012053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Mosquito-borne arboviruses are expanding their territory and elevating their infection prevalence due to the rapid climate change, urbanization, and increased international travel and global trade. Various significant arboviruses, including the dengue virus, Zika virus, Chikungunya virus, and yellow fever virus, are all reliant on the same primary vector, Aedes aegypti. Consequently, the occurrence of arbovirus coinfection in mosquitoes is anticipated. Arbovirus coinfection in mosquitoes has two patterns: simultaneous and sequential. Numerous studies have demonstrated that simultaneous coinfection of arboviruses in mosquitoes is unlikely to exert mutual developmental influence on these viruses. However, the viruses' interplay within a mosquito after the sequential coinfection seems intricated and not well understood. METHODOLOGY/PRINCIPAL FINDINGS We conducted experiments aimed at examining the phenomenon of arbovirus sequential coinfection in both mosquito cell line (C6/36) and A. aegypti, specifically focusing on dengue virus (DENV, serotype 2) and Zika virus (ZIKV). We firstly observed that DENV and ZIKV can sequentially infect mosquito C6/36 cell line, but the replication level of the subsequently infected ZIKV was significantly suppressed. Similarly, A. aegypti mosquitoes can be sequentially coinfected by these two arboviruses, regardless of the order of virus exposure. However, the replication, dissemination, and the transmission potential of the secondary virus were significantly inhibited. We preliminarily explored the underlying mechanisms, revealing that arbovirus-infected mosquitoes exhibited activated innate immunity, disrupted lipid metabolism, and enhanced RNAi pathway, leading to reduced susceptibility to the secondary arbovirus infections. CONCLUSIONS/SIGNIFICANCE Our findings suggest that, in contrast to simultaneous arbovirus coinfection in mosquitoes that can promote the transmission and co-circulation of these viruses, sequential coinfection appears to have limited influence on arbovirus transmission dynamics. However, it is important to note that more experimental investigations are needed to refine and expand upon this conclusion.
Collapse
Affiliation(s)
- Jiameng Peng
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Meichun Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Gang Wang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Dongjing Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Xiaoying Zheng
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Yongjun Li
- Department of Pathogen Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Vandana V, Dong S, Sheth T, Sun Q, Wen H, Maldonado A, Xi Z, Dimopoulos G. Wolbachia infection-responsive immune genes suppress Plasmodium falciparum infection in Anopheles stephensi. PLoS Pathog 2024; 20:e1012145. [PMID: 38598552 PMCID: PMC11034644 DOI: 10.1371/journal.ppat.1012145] [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: 01/04/2024] [Revised: 04/22/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
Wolbachia, a maternally transmitted symbiotic bacterium of insects, can suppress a variety of human pathogens in mosquitoes, including malaria-causing Plasmodium in the Anopheles vector. However, the mechanistic basis of Wolbachia-mediated Plasmodium suppression in mosquitoes is not well understood. In this study, we compared the midgut and carcass transcriptomes of stably infected Anopheles stephensi with Wolbachia wAlbB to uninfected mosquitoes in order to discover Wolbachia infection-responsive immune genes that may play a role in Wolbachia-mediated anti-Plasmodium activity. We show that wAlbB infection upregulates 10 putative immune genes and downregulates 14 in midguts, while it upregulates 31 putative immune genes and downregulates 15 in carcasses at 24 h after blood-fed feeding, the time at which the Plasmodium ookinetes are traversing the midgut tissue. Only a few of these regulated immune genes were also significantly differentially expressed between Wolbachia-infected and non-infected midguts and carcasses of sugar-fed mosquitoes. Silencing of the Wolbachia infection-responsive immune genes TEP 4, TEP 15, lysozyme C2, CLIPB2, CLIPB4, PGRP-LD and two novel genes (a peritrophin-44-like gene and a macro domain-encoding gene) resulted in a significantly greater permissiveness to P. falciparum infection. These results indicate that Wolbachia infection modulates mosquito immunity and other processes that are likely to decrease Anopheles permissiveness to Plasmodium infection.
Collapse
Affiliation(s)
- Vandana Vandana
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Tanaya Sheth
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Qiang Sun
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Han Wen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, United States of America
| | - Amanda Maldonado
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Zhiyong Xi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, 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
| |
Collapse
|
8
|
Garrigós M, Ylla G, Martínez-de la Puente J, Figuerola J, Ruiz-López MJ. Two avian Plasmodium species trigger different transcriptional responses on their vector Culex pipiens. Mol Ecol 2023:e17240. [PMID: 38108558 DOI: 10.1111/mec.17240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Malaria is a mosquito-borne disease caused by protozoans of the genus Plasmodium that affects both humans and wildlife. The fitness consequences of infections by avian malaria are well known in birds, however, little information exists on its impact on mosquitoes. Here we study how Culex pipiens mosquitoes transcriptionally respond to infection by two different Plasmodium species, P. relictum and P. cathemerium, differing in their virulence (mortality rate) and transmissibility (parasite presence in exposed mosquitoes' saliva). We studied the mosquito response to the infection at three critical stages of parasite development: the formation of ookinetes at 24 h post-infection (hpi), the release of sporozoites into the hemocoel at 10 days post-infection (dpi), and the storage of sporozoites in the salivary glands at 21 dpi. For each time point, we characterized the gene expression of mosquitoes infected with each P. relictum and P. cathemerium and mosquitoes fed on an uninfected bird and, subsequently, compared their transcriptomic responses. Differential gene expression analysis showed that most transcriptomic changes occurred during the early infection stage (24 hpi), especially when comparing P. relictum and P. cathemerium-infected mosquitoes. Differentially expressed genes in mosquitoes infected with each species were related mainly to the metabolism of the immune response, trypsin, and other serine-proteases. We conclude that these differences in response may partly play a role in the differential virulence and transmissibility previously observed between P. relictum and P. cathemerium in Cx. pipiens.
Collapse
Affiliation(s)
- Marta Garrigós
- Department of Parasitology, University of Granada, Granada, Spain
| | - Guillem Ylla
- Bioinformatics and Genome Biology Lab, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Josué Martínez-de la Puente
- Department of Parasitology, University of Granada, Granada, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Jordi Figuerola
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| | - María José Ruiz-López
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| |
Collapse
|
9
|
Boehm EC, Jaeger AS, Ries HJ, Castañeda D, Weiler AM, Valencia CC, Weger-Lucarelli J, Ebel GD, O’Connor SL, Friedrich TC, Zamanian M, Aliota MT. Wolbachia-mediated resistance to Zika virus infection in Aedes aegypti is dominated by diverse transcriptional regulation and weak evolutionary pressures. PLoS Negl Trop Dis 2023; 17:e0011674. [PMID: 37782672 PMCID: PMC10569609 DOI: 10.1371/journal.pntd.0011674] [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: 06/26/2023] [Revised: 10/12/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
A promising candidate for arbovirus control and prevention relies on replacing arbovirus-susceptible Aedes aegypti populations with mosquitoes that have been colonized by the intracellular bacterium Wolbachia and thus have a reduced capacity to transmit arboviruses. This reduced capacity to transmit arboviruses is mediated through a phenomenon referred to as pathogen blocking. Pathogen blocking has primarily been proposed as a tool to control dengue virus (DENV) transmission, however it works against a range of viruses, including Zika virus (ZIKV). Despite years of research, the molecular mechanisms underlying pathogen blocking still need to be better understood. Here, we used RNA-seq to characterize mosquito gene transcription dynamics in Ae. aegypti infected with the wMel strain of Wolbachia that are being released by the World Mosquito Program in Medellín, Colombia. Comparative analyses using ZIKV-infected, uninfected tissues, and mosquitoes without Wolbachia revealed that the influence of wMel on mosquito gene transcription is multifactorial. Importantly, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to pathogen blocking. Therefore, to understand the influence of Wolbachia on within-host ZIKV evolution, we characterized the genetic diversity of molecularly barcoded ZIKV virus populations replicating in Wolbachia-infected mosquitoes and found that within-host ZIKV evolution was subject to weak purifying selection and, unexpectedly, loose anatomical bottlenecks in the presence and absence of Wolbachia. Together, these findings suggest that there is no clear transcriptional profile associated with Wolbachia-mediated ZIKV restriction, and that there is no evidence for ZIKV escape from this restriction in our system.
Collapse
Affiliation(s)
- Emma C. Boehm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Hunter J. Ries
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - David Castañeda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Corina C. Valencia
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - James Weger-Lucarelli
- Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Gregory D. Ebel
- Colorado State University, Fort Collins, Colorado, United States of America
| | - Shelby L. O’Connor
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| |
Collapse
|
10
|
Hixson B, Huot L, Morejon B, Yang X, Nagy P, Michel K, Buchon N. The transcriptional response in mosquitoes distinguishes between fungi and bacteria but not Gram types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550663. [PMID: 37546902 PMCID: PMC10402080 DOI: 10.1101/2023.07.26.550663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Mosquitoes are prolific vectors of human pathogens; a clear and accurate understanding of the organization of their antimicrobial defenses is crucial for informing the development of transmission control strategies. The canonical infection response in insects, as described in the insect model Drosophila melanogaster , is pathogen type-dependent, with distinct stereotypical responses to Gram-negative bacteria and Gram-positive bacteria/fungi mediated by the activation of the Imd and Toll pathways, respectively. To determine whether this pathogen-specific discrimination is shared by mosquitoes, we used RNAseq to capture the genome-wide transcriptional response of Aedes aegypti and Anopheles gambiae ( s.l. ) to systemic infection with Gram-negative bacteria, Gram-positive bacteria, yeasts, and filamentous fungi, as well as challenge with heat-killed Gram-negative, Gram-positive, and fungal pathogens. From the resulting data, we found that Ae. aegypti and An. gambiae both mount a core response to all categories of infection, and this response is highly conserved between the two species with respect to both function and orthology. When we compared the transcriptomes of mosquitoes infected with different types of bacteria, we observed that the intensity of the transcriptional response was correlated with both the virulence and growth rate of the infecting pathogen. Exhaustive comparisons of the transcriptomes of Gram-negative-challenged versus Gram-positive-challenged mosquitoes yielded no difference in either species. In Ae. aegypti , however, we identified transcriptional signatures specific to bacterial infection and to fungal infection. The bacterial infection response was dominated by the expression of defensins and cecropins, while the fungal infection response included the disproportionate upregulation of an uncharacterized family of glycine-rich proteins. These signatures were also observed in Ae. aegypti challenged with heat-killed bacteria and fungi, indicating that this species can discriminate between molecular patterns that are specific to bacteria and to fungi.
Collapse
|
11
|
Boehm EC, Jaeger AS, Ries HJ, Castañeda D, Weiler AM, Valencia CC, Weger-Lucarelli J, Ebel GD, O’Connor SL, Friedrich TC, Zamanian M, Aliota MT. Wolbachia -mediated resistance to Zika virus infection in Aedes aegypti is dominated by diverse transcriptional regulation and weak evolutionary pressures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546271. [PMID: 37425681 PMCID: PMC10327090 DOI: 10.1101/2023.06.26.546271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A promising candidate for arbovirus control and prevention relies on replacing arbovirus-susceptible Aedes aegypti populations with mosquitoes that have been colonized by the intracellular bacterium Wolbachia and thus have a reduced capacity to transmit arboviruses. This reduced capacity to transmit arboviruses is mediated through a phenomenon referred to as pathogen blocking. Pathogen blocking has primarily been proposed as a tool to control dengue virus (DENV) transmission, however it works against a range of viruses, including Zika virus (ZIKV). Despite years of research, the molecular mechanisms underlying pathogen blocking still need to be better understood. Here, we used RNA-seq to characterize mosquito gene transcription dynamics in Ae. aegypti infected with the w Mel strain of Wolbachia that are being released by the World Mosquito Program in Medellín, Colombia. Comparative analyses using ZIKV-infected, uninfected tissues, and mosquitoes without Wolbachia revealed that the influence of w Mel on mosquito gene transcription is multifactorial. Importantly, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to pathogen blocking. Therefore, to understand the influence of Wolbachia on within-host ZIKV evolution, we characterized the genetic diversity of molecularly barcoded ZIKV virus populations replicating in Wolbachia -infected mosquitoes and found that within-host ZIKV evolution was subject to weak purifying selection and, unexpectedly, loose anatomical bottlenecks in the presence and absence of Wolbachia . Together, these findings suggest that there is no clear transcriptional profile associated with Wolbachia -mediated ZIKV restriction, and that there is no evidence for ZIKV escape from this restriction in our system. Author Summary When Wolbachia bacteria infect Aedes aegypti mosquitoes, they dramatically reduce the mosquitoes' susceptibility to infection with a range of arthropod-borne viruses, including Zika virus (ZIKV). Although this pathogen-blocking effect has been widely recognized, its mechanisms remain unclear. Furthermore, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to Wolbachia -mediated blocking. Here, we use host transcriptomics and viral genome sequencing to examine the mechanisms of ZIKV pathogen blocking by Wolbachia and viral evolutionary dynamics in Ae. aegypti mosquitoes. We find complex transcriptome patterns that do not suggest a single clear mechanism for pathogen blocking. We also find no evidence that Wolbachia exerts detectable selective pressures on ZIKV in coinfected mosquitoes. Together our data suggest that it may be difficult for ZIKV to evolve Wolbachia resistance, perhaps due to the complexity of the pathogen blockade mechanism.
Collapse
Affiliation(s)
- Emma C. Boehm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Hunter J. Ries
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - David Castañeda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Corina C. Valencia
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
| | | | | | - Shelby L. O’Connor
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, WI, United States
| | - Thomas C. Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| |
Collapse
|
12
|
Lapadula WJ, Juri Ayub M. Ribosome Inactivating Proteins in Insects: HGT, gene expression, and functional implications. Gene 2023:147547. [PMID: 37286020 DOI: 10.1016/j.gene.2023.147547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023]
Abstract
Ribosome-inactivating proteins (RIPs) are RNA N-glycosidases that depurinate an adenine residue in the conserved alpha-sarcin/ricin loop (SRL) of rRNA, inhibiting protein synthesis. Previously, we reported the existence of these toxins in insects, whose presence is restricted to mosquitoes from the Culicinae subfamily (e.g., Aedes aegypti) and whiteflies from the Aleyrodidae family (e.g., Bemisia tabaci). Both groups of genes are derived from two independent horizontal gene transfer (HGT) events and are evolved under purifying selection. Here, we report and characterize the occurrence of a third HGT event in the Sciaroidea superfamily, which supports the recurrent acquisition of RIP genes by insects. Transcriptomic experiments, available in databases, allowed us to describe the temporal and spatial expression profiles for these foreign genes in these organisms. Furthermore, we found that RIP expression is induced after infection with pathogens and provided, for the first time, transcriptomic evidence of parasite SRL depurination. This evidence suggests a possible role of these foreign genes as immune effectors in insects.
Collapse
Affiliation(s)
- Walter J Lapadula
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, IMIBIO-SL-CONICET and Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes, 950, D5700HHW San Luis, Argentina.
| | - Maximiliano Juri Ayub
- Instituto Multidisciplinario de Investigaciones Biológicas de San Luis, IMIBIO-SL-CONICET and Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de Los Andes, 950, D5700HHW San Luis, Argentina
| |
Collapse
|
13
|
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: 2.0] [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
|
14
|
Transcriptome Analysis of Response to Zika Virus Infection in Two Aedes albopictus Strains with Different Vector Competence. Int J Mol Sci 2023; 24:ijms24054257. [PMID: 36901688 PMCID: PMC10002152 DOI: 10.3390/ijms24054257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Zika virus (ZIKV), which is mainly transmitted by Aedes albopictus in temperate zones, can causes serious neurological disorders. However, the molecular mechanisms that influence the vector competence of Ae. albopictus for ZIKV are poorly understood. In this study, the vector competence of Ae. albopictus mosquitoes from Jinghong (JH) and Guangzhou (GZ) Cities of China were evaluated, and transcripts in the midgut and salivary gland tissues were sequenced on 10 days post-infection. The results showed that both Ae. albopictus JH and GZ strains were susceptible to ZIKV, but the GZ strain was more competent. The categories and functions of differentially expressed genes (DEGs) in response to ZIKV infection were quite different between tissues and strains. Through a bioinformatics analysis, a total of 59 DEGs that may affect vector competence were screened-among which, cytochrome P450 304a1 (CYP304a1) was the only gene significantly downregulated in both tissues of two strains. However, CYP304a1 did not influence ZIKV infection and replication in Ae. albopictus under the conditions set in this study. Our results demonstrated that the different vector competence of Ae. albopictus for ZIKV may be determined by the transcripts in the midgut and salivary gland, which will contribute to understanding ZIKV-mosquito interactions and develop arbovirus disease prevention strategies.
Collapse
|
15
|
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: 1.0] [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
|
16
|
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
|
17
|
Sneed SD, Dwivedi SB, DiGate C, Denecke S, Povelones M. Aedes aegypti Malpighian tubules are immunologically activated following systemic Toll activation. Parasit Vectors 2022; 15:469. [PMID: 36522779 PMCID: PMC9753289 DOI: 10.1186/s13071-022-05567-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Canine heartworm is a widespread and potentially fatal mosquito-borne disease caused by infections with the parasitic nematode, Dirofilaria immitis. We have previously shown that systemic activation of the Toll immune pathway via silencing of the negative regulator Cactus in Aedes aegypti blocks parasite development in the Malpighian tubules (MT), the mosquito renal organ. However, it was not established whether the MT were directly responding to Toll activation or were alternatively responding to upregulated proteins or other changes to the hemolymph driven by other tissues. Distinguishing these possibilities is crucial for developing more precise strategies to block D. immitis while potentially avoiding the fitness cost to the mosquito associated with Cactus silencing. METHODS This study defines the transcriptional response of the MT and changes to the hemolymph proteome of Ae. aegypti after systemic Toll activation via intra-thoracic injection of double-stranded Cactus (dsCactus) RNA. RESULTS Malpighian tubules significantly increased expression of the Toll pathway target genes that significantly overlapped expression changes occurring in whole mosquitoes. A significant overlap between the transcriptional response of the MT and proteins upregulated in the hemolymph was also observed. CONCLUSIONS Our data show that MT are capable of RNA interference-mediated gene silencing and directly respond to dsCactus treatment by upregulating targets of the canonical Toll pathway. Although not definitive, the strong correspondence between the MT transcriptional response and the hemolymph proteomic responses provides evidence that the MT may contribute to mosquito humoral immunity.
Collapse
Affiliation(s)
- Sarah D. Sneed
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sutopa B. Dwivedi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Cameron DiGate
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Shane Denecke
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Michael Povelones
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
18
|
Guo Y, Guo J, Li Y. Wolbachia wPip Blocks Zika Virus Transovarial Transmission in Aedes albopictus. Microbiol Spectr 2022; 10:e0263321. [PMID: 35894613 PMCID: PMC9603370 DOI: 10.1128/spectrum.02633-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/14/2022] [Indexed: 01/04/2023] Open
Abstract
Wolbachia is being developed as a biological tool to suppress mosquito populations and/or interfere with their transmitted viruses. Adult males with an artificial Wolbachia infection have been released, successfully yielding population suppression in multiple field trials. The main characteristic of the artificial Wolbachia-infected mosquitoes used in the suppression program is the lower vector competence than that in native infected/uninfected mosquitoes in horizontal and vertical transmission. Our previous studies have demonstrated that the Aedes albopictus HC line infected with a trio of Wolbachia strains exhibited almost complete blockade of dengue virus (DENV) and Zika virus (ZIKV) in horizontal and vertical transmission. However, the extent to which Wolbachia inhibits virus transovarial transmission is unknown since no studies have been performed to determine whether Wolbachia protects ovarian cells against viral infection. Here, we employed ovarian cells of the Ae. albopictus GUA (a wild-type mosquito line superinfected with two native Wolbachia strains, wAlbA and wAlbB), HC, and GT lines (tetracycline-cured, Wolbachia-uninfected mosquitoes), which exhibit key traits, and compared them to better understand how Wolbachia inhibits ZIKV transovarial transmission. Our results showed that the infection rate of adult GT progeny was significantly higher than that of GUA progeny during the first and second gonotrophic cycles. In contrast, the infection rates of adult GT and GUA progeny were not significantly different during the third gonotrophic cycle. All examined adult HC progeny from three gonotrophic cycles were negative for ZIKV infection. A strong negative linear correlation existed between Wolbachia density and ZIKV load in the ovaries of mosquitoes. Although there is no obvious coexistence area in the ovaries for Wolbachia and ZIKV, host immune responses may play a role in Wolbachia blocking ZIKV expansion and maintenance in the ovaries of Ae. albopictus. These results will aid in understanding Wolbachia-ZIKV interactions in mosquitoes. IMPORTANCE Area-wide application of Wolbachia to suppress mosquito populations and their transmitted viruses has achieved success in multiple countries. However, the mass release of Wolbachia-infected male mosquitoes involves a potential risk of accidentally releasing fertile females. In this study, we employed ovarian cells of the Ae. albopictus GUA, HC, and GT lines, which exhibit key traits, and compared them to better understand how Wolbachia inhibits ZIKV transovarial transmission. Our results showed an almost complete blockade of ZIKV transmission in HC female mosquitoes. Wolbachia in natively infected GUA mosquitoes negative affected ZIKV, and this interference was shown by slightly lower loads than those in HC mosquitoes. Overall, our work helps show how Wolbachia blocks ZIKV expansion and maintenance in the ovaries of Ae. albopictus and aids in understanding Wolbachia-ZIKV interactions in mosquitoes.
Collapse
Affiliation(s)
- Yan Guo
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Science, Guangzhou, Guangdong, China
| | - Jiatian Guo
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yifeng Li
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Science, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Wang M, Wang Y, Chang M, Wang X, Shi Z, Raikhel AS, Zou Z. Ecdysone signaling mediates the trade-off between immunity and reproduction via suppression of amyloids in the mosquito Aedes aegypti. PLoS Pathog 2022; 18:e1010837. [PMID: 36137163 PMCID: PMC9531809 DOI: 10.1371/journal.ppat.1010837] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/04/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
The balance between immunity and reproduction is essential for many key physiological functions. We report that to maintain an optimal fertility, 20-hydroxyecdysone (20E) and the ecdysone receptor (EcR) downregulate the immune deficiency (IMD) pathway during the post blood meal phase (PBM) of the Aedes aegypti reproductive cycle. RNA interference-mediated depletion of EcR elicited an increased expression of the IMD pathway components, and these mosquitoes were more resistant to infection by Gram-negative bacteria. Moreover, 20E and EcR recruit Pirk-like, the mosquito ortholog of Drosophila melanogaster Pirk. CRISPR-Cas9 knockout of Pirk-like has shown that it represses the IMD pathway by interfering with IMD-mediated formation of amyloid aggregates. 20E and EcR disruption of the amyloid formation is pivotal for maintaining normal yolk protein production and fertility. Additionally, 20E and its receptor EcR directly induce Pirk-like to interfere with cRHIM-mediated formation of amyloid. Our study highlights the vital role of 20E in governing the trade-off between immunity and reproduction. Pirk-like might be a potential target for new methods to control mosquito reproduction and pathogen transmission.
Collapse
Affiliation(s)
- Mao Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yanhong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Mengmeng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xueli Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zuokun Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Alexander S. Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
- * E-mail: (ASR); (ZZ)
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, People’s Republic of China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- * E-mail: (ASR); (ZZ)
| |
Collapse
|
20
|
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: 1.5] [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
|
21
|
Rosendo Machado S, van der Most T, Miesen P. Genetic determinants of antiviral immunity in dipteran insects - Compiling the experimental evidence. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104010. [PMID: 33476667 DOI: 10.1016/j.dci.2021.104010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
The genetic basis of antiviral immunity in dipteran insects is extensively studied in Drosophila melanogaster and advanced technologies for genetic manipulation allow a better characterization of immune responses also in non-model insect species. Especially, immunity in vector mosquitoes is recently in the spotlight, due to the medical impact that these insects have by transmitting viruses and other pathogens. Here, we review the current state of experimental evidence that supports antiviral functions for immune genes acting in different cellular pathways. We discuss the well-characterized RNA interference mechanism along with the less well-defined JAK-STAT, Toll, and IMD signaling pathways. Furthermore, we highlight the initial evidence for antiviral activity observed for the autophagy pathway, transcriptional pausing, as well as piRNA production from endogenous viral elements. We focus our review on studies from Drosophila and mosquito species from the lineages Aedes, Culex, and Anopheles, which contain major vector species responsible for virus transmission.
Collapse
Affiliation(s)
- Samara Rosendo Machado
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Tom van der Most
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands
| | - Pascal Miesen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, the Netherlands.
| |
Collapse
|
22
|
Abstract
Zika virus (ZIKV; Flaviviridae) is a devastating virus transmitted to humans by the mosquito Aedes aegypti. The interaction of the virus with the mosquito vector is poorly known. The double-stranded RNA (dsRNA)-mediated interruption or activation of immunity-related genes in the Toll, IMD, JAK-STAT, and short interfering RNA (siRNA) pathways did not affect ZIKV infection in A. aegypti. Transcriptome-based analysis indicated that most immunity-related genes were upregulated in response to ZIKV infection, including leucine-rich immune protein (LRIM) genes. Further, there was a significant increment in the ZIKV load in LRIM9-, LRIM10A-, and LIRM10B-silenced A. aegypti, suggesting their function in modulating viral infection. Further, gene function enrichment analysis revealed that viral infection increased global ribosomal activity. Silencing of RpL23 and RpL27, two ribosomal large subunit genes, increased mosquito resistance to ZIKV infection. In vitro fat body culture assay revealed that the expression of RpL23 and RpL27 was responsive to the Juvenile hormone (JH) signaling pathway. These two genes were transcriptionally regulated by JH and its receptor methoprene-tolerant (Met) complex. Silencing of Met also inhibited ZIKV infection in A. aegypti. This suggests that ZIKV enhances ribosomal activity through JH regulation to promote infection in mosquitoes. Together, these data reveal A. aegypti immune responses to ZIKV and suggest a control strategy that reduces ZIKV transmission by modulating host factors. IMPORTANCE Most flaviviruses are transmitted between hosts by arthropod vectors such as mosquitoes. Since therapeutics or vaccines are lacking for most mosquito-borne diseases, reducing the mosquito vector competence is an effective way to decrease disease burden. We used high-throughput sequencing technology to study the interaction between mosquito Aedes aegypti and ZIKV. Leucine-rich immune protein (LRIM) genes were involved in the defense in response to viral infection. In addition, RNA interference (RNAi) silencing of RpL23 and RpL27, two JH-regulated ribosomal large subunit genes, suppressed ZIKV infection in A. aegypti. These results suggest a novel control strategy that could block the transmission of ZIKV.
Collapse
|
23
|
Russell TA, Ayaz A, Davidson AD, Fernandez-Sesma A, Maringer K. Imd pathway-specific immune assays reveal NF-κB stimulation by viral RNA PAMPs in Aedes aegypti Aag2 cells. PLoS Negl Trop Dis 2021; 15:e0008524. [PMID: 33591970 PMCID: PMC7909628 DOI: 10.1371/journal.pntd.0008524] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/26/2021] [Accepted: 01/23/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The mosquito Aedes aegypti is a major vector for the arthropod-borne viruses (arboviruses) chikungunya, dengue, yellow fever and Zika viruses. Vector immune responses pose a major barrier to arboviral transmission, and transgenic insects with altered immunity have been proposed as tools for reducing the global public health impact of arboviral diseases. However, a better understanding of virus-immune interactions is needed to progress the development of such transgenic insects. Although the NF-κB-regulated Toll and 'immunodeficiency' (Imd) pathways are increasingly thought to be antiviral, relevant pattern recognition receptors (PRRs) and pathogen-associated molecular patterns (PAMPs) remain poorly characterised in A. aegypti. METHODOLOGY/PRINCIPLE FINDINGS We developed novel RT-qPCR and luciferase reporter assays to measure induction of the Toll and Imd pathways in the commonly used A. aegypti-derived Aag2 cell line. We thus determined that the Toll pathway is not inducible by exogenous stimulation with bacterial, viral or fungal stimuli in Aag2 cells under our experimental conditions. We used our Imd pathway-specific assays to demonstrate that the viral dsRNA mimic poly(I:C) is sensed by the Imd pathway, likely through intracellular and extracellular PRRs. The Imd pathway was also induced during infection with the model insect-specific virus cricket paralysis virus (CrPV). CONCLUSIONS/SIGNIFICANCE Our demonstration that a general PAMP shared by many arboviruses is sensed by the Imd pathway paves the way for future studies to determine how viral RNA is sensed by mosquito PRRs at a molecular level. Our data also suggest that studies measuring inducible immune pathway activation through antimicrobial peptide (AMP) expression in Aag2 cells should be interpreted cautiously given that the Toll pathway is not responsive under all experimental conditions. With no antiviral therapies and few effective vaccines available to treat arboviral diseases, our findings provide new insights relevant to the development of transgenic mosquitoes as a means of reducing arbovirus transmission.
Collapse
Affiliation(s)
- Tiffany A. Russell
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Andalus Ayaz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Andrew D. Davidson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kevin Maringer
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
- The Pirbright Institute, Pirbright, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Wang L, Liu C, Geng X. Identify immune-related genes of adzuki bean weevil (Callosobruchus chinensis) in response to bacteria challenge by transcriptome analysis. Microb Pathog 2021; 151:104749. [PMID: 33484809 DOI: 10.1016/j.micpath.2021.104749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Callosobruchus chinensis is one of the important postharvest pests in legume growing areas. Bacterial pesticide is a potential alternative method to control storage pests. However, the effect of these pathogen bacteria on storage pests, and the molecular mechanisms of insect response remain to be to investigated. RESULTS Using the next generation sequencing technology, we established a transcriptomic library for C. chinensis larvae in response to Escherichia coli. Total of 355 differential expressed genes (DEGs) were identified, which 178 DEGs were upregulated, and 177 DEGs were downregulated compared to control group. To validate the RNA-seq analysis, 20 DEGs and 14 immune-related genes were selected to perform quantitative polymerase chain reaction (RT-qPCR). These immune-related genes were involved in recognition (peptidoglycan recognition proteins), signal transduction (fibrinogen-related proteins, serine proteinases and NF-κB), and execution effectors (phenoloxidase, defensin, attacin, and antimicrobial peptide). In addition, genes that encode digestive and respiratory enzymes were altered in C. chinensis larvae in response to infection. Some genes that involved in juvenile hormone and insulin pathway appeared to express differentially, suggesting that pathogen infection might lead to developmental arrest. Furthermore, iron homeostasis and chitin metabolism appeared significantly altered after infection. CONCLUSION In this study, we characterized the immune response of C. chinensis larvae in response to E. coli using RNA-seq, from pathogen recognition, signal transduction, to execution. Some other identified genes were involved in iron homeostasis, respiration, and digestion. A better understanding of molecular response of beetle to pathogen will facilitate us to develop an available strategy to control storage pests.
Collapse
Affiliation(s)
- Lei Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, 212100, PR China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Chang Liu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, 212100, PR China
| | - Xueqing Geng
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| |
Collapse
|
25
|
He YJ, Lu G, Qi YH, Zhang Y, Zhang XD, Huang HJ, Zhuo JC, Sun ZT, Yan F, Chen JP, Zhang CX, Li JM. Activation of Toll Immune Pathway in an Insect Vector Induced by a Plant Virus. Front Immunol 2021; 11:613957. [PMID: 33488623 PMCID: PMC7821435 DOI: 10.3389/fimmu.2020.613957] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/01/2020] [Indexed: 11/17/2022] Open
Abstract
The Toll pathway plays an important role in defense against infection of various pathogenic microorganisms, including viruses. However, current understanding of Toll pathway was mainly restricted in mammal and some model insects such as Drosophila and mosquitoes. Whether plant viruses can also activate the Toll signaling pathway in vector insects is still unknown. In this study, using rice stripe virus (RSV) and its insect vector (small brown planthopper, Laodelphax striatellus) as a model, we found that the Toll pathway was activated upon RSV infection. In comparison of viruliferous and non-viruliferous planthoppers, we found that four Toll pathway core genes (Toll, Tube, MyD88, and Dorsal) were upregulated in viruliferous planthoppers. When the planthoppers infected with RSV, the expressions of Toll and MyD88 were rapidly upregulated at the early stage (1 and 3 days post-infection), whereas Dorsal was upregulated at the late stage (9 days post-infection). Furthermore, induction of Toll pathway was initiated by interaction between a Toll receptor and RSV nucleocapsid protein (NP). Knockdown of Toll increased the proliferation of RSV in vector insect, and the dsToll-treated insects exhibited higher mortality than that of dsGFP-treated ones. Our results provide the first evidence that the Toll signaling pathway of an insect vector is potentially activated through the direct interaction between Toll receptor and a protein encoded by a plant virus, indicating that Toll immune pathway is an important strategy against plant virus infection in an insect vector.
Collapse
Affiliation(s)
- Yu-Juan He
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Gang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Yu-Hua Qi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Yan Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Xiao-Di Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Hai-Jian Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Ji-Chong Zhuo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Zong-Tao Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Fei Yan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jian-Ping Chen
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China.,State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Chuan-Xi Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jun-Min Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MOA of China and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| |
Collapse
|
26
|
Chang MM, Wang YH, Yang QT, Wang XL, Wang M, Raikhel AS, Zou Z. Regulation of antimicrobial peptides by juvenile hormone and its receptor, Methoprene-tolerant, in the mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 128:103509. [PMID: 33264664 DOI: 10.1016/j.ibmb.2020.103509] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 11/09/2020] [Accepted: 11/24/2020] [Indexed: 06/12/2023]
Abstract
The trade-off between reproduction and immunity has been established for a number of insect species. However, the regulatory mechanisms governing this event is not well understood. In the mosquito Aedes aegypti, the vector of dangerous human arboviral diseases, juvenile hormone (JH) is required for the female post-eclosion development and reproductive maturation. In this study, we have revealed the JH negative effect on the expression of immunity-related genes, such as antimicrobial peptides (AMPs), during the post-eclosion phase of the female mosquito gonadotrophic reproductive cycle. Mosquitoes treated with JH became more sensitive to microbial infection. Mosquitoes subjected to the RNA interference knockdown (RNAi) of the JH receptor, Methoprene-tolerant (Met), showed increased expression of several AMP genes. Met binds to the E-box-like recognition motifs in the regulatory region of the diptericin (Dpt) gene, indicating that JH can suppress the Dpt gene expression through its receptor Met. Hence, JH is involved in the modulation of immune responses during the post-eclosion phase of reproduction. The RNAi knockdown of the peptidoglycan recognition protein (PGRP-LC) led to a significant reduction of the Dpt transcript level, indicating the PGRP-LC activating role on this AMP gene. Thus, Dpt appeared to be under the dual regulation of both the JH and the immune deficiency (IMD) signaling pathways. Our study provides a better understanding of how JH regulates insect immunity in adult mosquitoes.
Collapse
Affiliation(s)
- Meng-Meng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yan-Hong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Tai Yang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xue-Li Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mao Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Alexander S Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, CA, 92521, USA
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, 311300, China.
| |
Collapse
|
27
|
Fernando DD, Fischer K. Proteases and pseudoproteases in parasitic arthropods of clinical importance. FEBS J 2020; 287:4284-4299. [PMID: 32893448 DOI: 10.1111/febs.15546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
Parasitic arthropods feed on blood or skin tissue and share comparable repertoires of proteases involved in haematophagy, digestion, egg development and immunity. While proteolytically active proteases of multiple classes dominate, an increasing number of pseudoproteases have been discovered that have no proteolytic function but are pharmacologically active biomolecules, evolved to carry out alternative functions as regulatory, antihaemostatic, anti-inflammatory or immunomodulatory compounds. In this review, we provide an overview of proteases and pseudoproteases from clinically important arthropod parasites. Many of these act in central biological pathways of parasite survival and host-parasite interaction and may be potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Deepani Darshika Fernando
- Cell and Molecular Biology Department, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| | - Katja Fischer
- Cell and Molecular Biology Department, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| |
Collapse
|
28
|
Activation of the Toll pathway in Aedes aegypti blocks the development of emerging third-stage larvae of drug-resistant Dirofilaria immitis. Vet Parasitol 2020; 282:109100. [PMID: 32460109 DOI: 10.1016/j.vetpar.2020.109100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 01/02/2023]
Abstract
Dirofilaria immitis is the globally distributed agent of heartworm disease. Infection in canines causes debilitating disease that can be fatal if left untreated. Macrocyclic lactones can prevent heartworm disease in dogs, cats and ferrets by killing larvae before they develop into adult worms in the pulmonary artery. However, administration of prophylactic drugs to wild canids to prevent D. immitis infection is not feasible. Furthermore, a vaccine against heartworm is currently unavailable and drug resistant D. immitis have been identified, highlighting the need for new strategies to prevent parasite transmission. We recently established a method to block development of emerging third-stage larvae (eL3) from the mosquito Aedes aegypti by over-activating the Toll pathway, one of the major innate immune signaling pathways in mosquitoes. Our previous study used a drug-sensitive strain of D. immitis and it remains unknown if the strategy is effective against different D. immitis genotypes and, more importantly, if it would work against drug-resistant genotypes. The purpose of this study was to determine whether Toll pathway activation is capable of blocking eL3 development of D. immitis strains that are resistant to macrocyclic lactones. We infected mosquitoes with two independent strains of D. immitis previously confirmed as being resistant to macrocyclic lactones, and then activated Toll signaling by RNAi-mediated silencing of the pathway inhibitor, IκB/Cactus, and quantitatively measured eL3 development. Similar to the drug-sensitive strain, eL3 were strongly reduced by Toll activation in both drug-resistant strains. Furthermore, similar to the drug-sensitive strain, the reduction of eL3 in both drug-resistant strains suggests a defect in larval invasion of, or development in, the Malpighian tubules - the organ in the mosquito to which microfilariae migrate after ingestion and where the larvae undergo several developmental molts. In summary, Toll pathway activation blocks the development of three distinct D. immitis genotypes, including two different drug-resistant genotypes. If this strategy can be applied to heartworm vectors in the field, it may help reduce the spread of disease and is not predicted to favor the spread of drug resistance.
Collapse
|
29
|
Ng TH, Kurtz J. Dscam in immunity: A question of diversity in insects and crustaceans. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 105:103539. [PMID: 31734281 DOI: 10.1016/j.dci.2019.103539] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/07/2019] [Accepted: 11/09/2019] [Indexed: 06/10/2023]
Abstract
In insects and crustaceans, thousands of Down syndrome cell adhesion molecules (Dscam) can be generated by alternative splicing of variable exons from a single-locus gene, Dscam-hv. This extraordinarily versatile gene (38,016 protein isoforms produced in Drosophila) was first proposed to be involved in exon guidance and subsequently implicated in immunity as a hypervariable immune molecule. Almost 20 y after discovery of Dscam-hv, there have been many studies in insects and crustaceans regarding roles of Dscam in immunity, with many similarities and concurrently, many differences. Here, we review the current status of Dscam-hv, presented as a comparison of similarities and differences in insects and crustaceans and discuss hypotheses of Dscam functions in immunity.
Collapse
Affiliation(s)
- Tze Hann Ng
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany
| | - Joachim Kurtz
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149, Münster, Germany.
| |
Collapse
|
30
|
Araujo RV, Feitosa-Suntheimer F, Gold AS, Londono-Renteria B, Colpitts TM. One-step RT-qPCR assay for ZIKV RNA detection in Aedes aegypti samples: a protocol to study infection and gene expression during ZIKV infection. Parasit Vectors 2020; 13:128. [PMID: 32171303 PMCID: PMC7071672 DOI: 10.1186/s13071-020-4002-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 02/29/2020] [Indexed: 12/22/2022] Open
Abstract
Background Zika virus (ZIKV) is transmitted to humans during the bite of an infected mosquito. In a scenario of globalization and climate change, the frequency of outbreaks has and will increase in areas with competent vectors, revealing a need for continuous improvement of ZIKV detection tools in vector populations. A simple, rapid and sensitive assay for viral detection is quantitative reverse transcription polymerase chain reaction (qRT-PCR), yet oligos optimized for ZIKV detection in mammalian cells and samples have repeatedly shown high background when used on mosquito ribonucleic acid (RNA). In this paper, we present a one-step qRT-PCR protocol that allows for the detection of ZIKV in mosquitoes and for the evaluation of gene expression from the same mosquito sample and RNA. This assay is a less expensive qRT-PCR approach than that most frequently used in the literature and has a much lower background, allowing confident detection. Methods Our new oligo design to detect ZIKV RNA included in silico analysis of both viral and mosquito (Ae. aegypti and Ae. albopictus) genomes, targeting sequences conserved between Asian and African ZIKV lineages, but not matching Aedes genomes. This assay will allow researchers to avoid nonspecific amplification in insect samples due to viral integration into the mosquito genome, a phenomenon known to happen in wild and colonized populations of mosquitoes. Standard curves constructed with in vitro transcribed ZIKV RNA were used to optimize the sensitivity, efficiency and reproducibility of the assay. Results Finally, the assay was used with success to detect both ZIKV RNA in infected mosquitoes and to detect expression of the Defensin A gene, an antimicrobial peptide (AMP) involved in Aedes aegypti immune response to virus infection. Conclusions The experimental approach to detect ZIKV RNA in Aedes aegypti presented here has demonstrated to be specific, sensitive and reliable, and additionally it allows for the analysis of mosquito gene expression during ZIKV infection.![]()
Collapse
Affiliation(s)
- Ricardo Vieira Araujo
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA.,Climate Division, Ministry of Science, Technology, Innovations and Communications, Brasilia, DF, Brazil
| | - Fabiana Feitosa-Suntheimer
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Alexander S Gold
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | | | - Tonya M Colpitts
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA. .,National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA.
| |
Collapse
|
31
|
Wang JY, Zhang H, Siemann E, Ji XY, Chen YJ, Wang Y, Jiang JX, Wan NF. Immunity of an insect herbivore to an entomovirus is affected by different host plants. PEST MANAGEMENT SCIENCE 2020; 76:1004-1010. [PMID: 31489764 DOI: 10.1002/ps.5609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 05/28/2023]
Abstract
BACKGROUND Interactions between herbivorous insects and entomoviruses may depend on host plant, perhaps mediated through changes in herbivore innate immunity. RESULTS Caterpillars (Spodoptera exigua) fed Glycine max had high viral loads and low melanization rates together with low melanization enzyme [PO, DDC, TH] activities and gene expressions. Caterpillars fed Ipomoea aquatica had low viral loads and high melanization, gene activities and gene expressions while those fed Brassica oleracea or artificial diet had intermediate levels of each. Melanization rates were negatively correlated with viral loads and positively correlated with activity and expression of each of the three enzymes. Some diet effects on enzymes were constitutive because the same diets led to low (G. max) or high (I. aquatica) melanization related gene activities and expressions without infection. CONCLUSION Diet influences the interactions between insect herbivores and viruses by shaping the innate immune response both at the onset of infection and afterwards as viral loads accumulate over a period of days. In addition, diets that lead to low viral loads are associated with high activities and gene expressions of a variety of melanization related enzymes suggesting a common causative mechanism. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jin-Yan Wang
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| | - Hao Zhang
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| | - Evan Siemann
- Department of Biosciences, Rice University, Houston, TX, USA
| | - Xiang-Yun Ji
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| | - Yi-Juan Chen
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| | - Yi Wang
- Laboratory of Ecology and Evolutionary Biology, Yunnan University, Kunming, China
| | - Jie-Xian Jiang
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| | - Nian-Feng Wan
- Eco-environmental Protection Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Protected Horticultural Technology, Shanghai Engineering Research Centre of Low-carbon Agriculture, Shanghai, China
| |
Collapse
|
32
|
Edgerton EB, McCrea AR, Berry CT, Kwok JY, Thompson LK, Watson B, Fuller EM, Nolan TJ, Lok JB, Povelones M. Activation of mosquito immunity blocks the development of transmission-stage filarial nematodes. Proc Natl Acad Sci U S A 2020; 117:3711-3717. [PMID: 32015105 PMCID: PMC7035481 DOI: 10.1073/pnas.1909369117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne helminth infections are responsible for a significant worldwide disease burden in both humans and animals. Accordingly, development of novel strategies to reduce disease transmission by targeting these pathogens in the vector are of paramount importance. We found that a strain of Aedes aegypti that is refractory to infection by Dirofilaria immitis, the agent of canine heartworm disease, mounts a stronger immune response during infection than does a susceptible strain. Moreover, activation of the Toll immune signaling pathway in the susceptible strain arrests larval development of the parasite, thereby decreasing the number of transmission-stage larvae. Notably, this strategy also blocks transmission-stage Brugia malayi, an agent of human lymphatic filariasis. Our data show that mosquito immunity can play a pivotal role in restricting filarial nematode development and suggest that genetically engineering mosquitoes with enhanced immunity will help reduce pathogen transmission.
Collapse
Affiliation(s)
- Elizabeth B Edgerton
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Abigail R McCrea
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Corbett T Berry
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Jenny Y Kwok
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Letitia K Thompson
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Brittany Watson
- Department of Clinical Sciences & Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | | | - Thomas J Nolan
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - James B Lok
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Michael Povelones
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104;
| |
Collapse
|
33
|
Diop F, Alout H, Diagne CT, Bengue M, Baronti C, Hamel R, Talignani L, Liegeois F, Pompon J, Morales Vargas RE, Nougairède A, Missé D. Differential Susceptibility and Innate Immune Response of Aedes aegypti and Aedes albopictus to the Haitian Strain of the Mayaro Virus. Viruses 2019; 11:v11100924. [PMID: 31601017 PMCID: PMC6832402 DOI: 10.3390/v11100924] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022] Open
Abstract
Mayaro (MAYV) is an emerging arthropod-borne virus belonging to the Alphavirus genus of the Togaviridae family. Although forest-dwelling Haemagogus mosquitoes have been considered as its main vector, the virus has also been detected in circulating Aedes ssp mosquitoes. Here we assess the susceptibility of Aedes aegypti and Aedes albopictus to infection with MAYV and their innate immune response at an early stage of infection. Aedes albopictus was more susceptible to infection with MAYV than Ae. aegypti. Analysis of transcript levels of twenty immunity-related genes by real-time PCR in the midgut of both mosquitoes infected with MAYV revealed increased expression of several immune genes, including CLIP-domain serine proteases, the anti-microbial peptides defensin A, E, cecropin E, and the virus inducible gene. The regulation of certain genes appeared to be Aedes species-dependent. Infection of Ae. aegypti with MAYV resulted in increased levels of myeloid differentiation2-related lipid recognition protein (ML26A) transcripts, as compared to Ae. albopictus. Increased expression levels of thio-ester-containing protein 22 (TEP22) and Niemann–Pick type C1 (NPC1) gene transcripts were observed in infected Ae. albopictus, but not Ae. aegypti. The differences in these gene expression levels during MAYV infection could explain the variation in susceptibility observed in both mosquito species.
Collapse
Affiliation(s)
- Fodé Diop
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Haoues Alout
- ASTRE, INRA CIRAD (UMR117), 34394 Montpellier, France.
| | | | - Michèle Bengue
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Cécile Baronti
- Unité des virus émergents, Aix Marseille Univ-IRD 190, Inserm 1207-IHU Méditerranée Infection, 13385 Marseille, France.
| | - Rodolphe Hamel
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Loïc Talignani
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Florian Liegeois
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Julien Pompon
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| | - Ronald E Morales Vargas
- Department of Medical Entomology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| | - Antoine Nougairède
- Unité des virus émergents, Aix Marseille Univ-IRD 190, Inserm 1207-IHU Méditerranée Infection, 13385 Marseille, France.
| | - Dorothée Missé
- MIVEGEC-IRD, Univ. Montpellier, CNRS, 34394 Montpellier, France.
| |
Collapse
|
34
|
Kong H, Dong C, Jing W, Zheng M, Tian Z, Hou Q, Wang C, Cheng Y, Zhang L, Jiang X, Luo L, Zhu S. Transcriptomic insight into antimicrobial peptide factors involved in the prophylactic immunity of crowded Mythimna separata larvae. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 98:34-41. [PMID: 30794832 DOI: 10.1016/j.dci.2019.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
Similar to pathogenic infection, a high population density alters insect prophylactic immunity. Antimicrobial peptides (AMPs) are known to play critical roles in an insect's humoral immune response to microbial infection. We applied RNA sequencing to investigate differential gene expression levels in fat body and hemocyte samples from larvae reared in high- (10 larvae per jar) and low-density (1 larva per jar) conditions; the samples exhibited density-dependent prophylaxis. A number of AMP molecule-related proteins were annotated for the first time from 145,439 assembled unigenes from M. separata larvae. The transcript levels of AMP molecules such as gloverin-, defensin-, cecropin-, lebocin- and attacin-related unigenes were increased with the prophylactic immunity of high-density larvae. The pattern recognition receptor peptidoglycan recognition protein (PGRP), a key protein in the synthesis of AMPs in IMD- and Toll pathway-related unigenes, was also upregulated in the larvae from the high-density group. The resultant transcriptomic database was validated by the transcript levels of four selected AMP genes quantified from the high- and low-density larval groups with quantitative real-time PCR. The antimicrobial activity against gram-positive Staphylococcus aureus and Bacillus subtilis and gram-negative Edwardsiella ictaluri and Vibrio anguillarum in the hemolymph of larvae from the high-density group was significantly higher than that of larvae from the low-density group. Our findings provide the first insight into the role of AMP genes in the mechanisms of density-dependent prophylaxis in M. separata and provide new insight into the control of M. separata with biopesticides.
Collapse
Affiliation(s)
- Hailong Kong
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China.
| | - Chuanlei Dong
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Wanghui Jing
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Minyuan Zheng
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Zhen Tian
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Qiuli Hou
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Cheng Wang
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| | - Yunxia Cheng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests; Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, China
| | - Lei Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests; Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, China
| | - Xingfu Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests; Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, China.
| | - Lizhi Luo
- State Key Laboratory for Biology of Plant Diseases and Insect Pests; Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, China
| | - Shude Zhu
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, China
| |
Collapse
|
35
|
Using genetic variation in Aedes aegypti to identify candidate anti-dengue virus genes. BMC Infect Dis 2019; 19:580. [PMID: 31272403 PMCID: PMC6611004 DOI: 10.1186/s12879-019-4212-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Transcriptomic profiling has generated extensive lists of genes that respond to viral infection in mosquitoes. These gene lists contain two types of genes; (1) those that are responsible for the insect's natural antiviral defense mechanisms, including some known innate immunity genes, and (2) genes whose change in expression may occur simply as a result of infection. As genetic modification tools for mosquitoes continue to improve, the opportunities to make refractory insects via allelic replacement or delivery of small RNAs that alter gene expression are expanding. Therefore, the ability to identify which genes in transcriptional profiles may have immune function has increasing value. Arboviruses encounter a range of mosquito tissues and physiologies as they traverse from the midgut to the salivary glands. While the midgut is well-studied as the primary tissue barrier, antiviral genes expressed in the subsequent tissues of the carcass offer additional candidates for second stage intervention in the mosquito body. METHODS Mosquito lines collected recently from field populations exhibit natural genetic variation for dengue virus susceptibility. We sought to use a modified full-sib breeding design to identify mosquito families that varied in their dengue viral load in their bodies post infection. RESULTS By delivering virus intrathoracically, we bypassed the midgut and focused on whole body responses in order to evaluate carcass-associated refractoriness. We tested 25 candidate genes selected for their appearance in multiple published transcriptional profiles and were able to identify 12 whose expression varied with susceptibility in the genetic families. CONCLUSIONS This method, using natural genetic variation, offers a simple means to screen and reduce candidate gene lists prior to carrying out more labor-intensive functional studies. The extracted RNA from the females across the families represents a storable resource that can be used to screen subsequent candidate genes in the future. The aspect of vector competence being assessed could be varied by focusing on different tissues or time points post infection.
Collapse
|
36
|
Aedes aegypti HPX8C modulates immune responses against viral infection. PLoS Negl Trop Dis 2019; 13:e0007287. [PMID: 30986216 PMCID: PMC6464178 DOI: 10.1371/journal.pntd.0007287] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
Mosquitoes act as vectors of numerous pathogens that cause human diseases. Dengue virus (DENV) transmitted by mosquito, Aedes aegypti, is responsible for dengue fever epidemics worldwide with a serious impact on human health. Currently, disease control mainly relies on vector targeted intervention strategies. Therefore, it is imperative to understand the molecular mechanisms underlying the innate immune response of mosquitoes against pathogens. In the present study, the expression profiles of immunity-related genes in the midgut responding to DENV infection by feeding were analyzed by transcriptome and quantitative real-time PCR. The level of Antimicrobial peptides (AMPs) increased seven days post-infection (d.p.i.), which could be induced by the Toll immune pathway. The expression of reactive oxygen species (ROS) genes, including antioxidant genes, such as HPX7, HPX8A, HPX8B, HPX8C were induced at one d.p.i. and peaked again at ten d.p.i. in the midgut. Interestingly, down-regulation of the antioxidant gene HPX8C by RNA interference led to reduction in the virus titer in the mosquito, probably due to the elevated levels of ROS. Application of a ROS inhibitor and scavenger molecules further established the role of oxygen free radicals in the modulation of the immune response to DENV infection. Overall, our comparative transcriptome analyses provide valuable information about the regulation of immunity related genes in the transmission vector in response to DENV infection. It further allows us to identify novel molecular mechanisms underlying the host-virus interaction, which might aid in the development of novel strategies to control mosquito-borne diseases. HPX8C is a heme-containing peroxidase, which can move reactive oxygen species (ROS) damage to the organism by reducing H2O2 to H2O. Previously, the peroxidase gene has been shown to modulate midgut immunity and regulate anti-malarial response in mosquitoes. In this study, the classical immune signaling pathways, Toll and IMD genes might be late responses against the viruses. HPX8C was demonstrated here to play a role in antiviral immunity against DENV infection in Ae. Aegypti mosquitoes. HPX8C expression was induced by DENV infection and continued to increase with an elevated virus titer. In HPX8C-depleted mosquitoes, the ROS level was found to be increased with a corresponding decrease in the DENV and ZIKV virus titer. Therefore, it was speculated that HPX8C mediated immune responses against the DENV in the mosquito in the late stage of viral infection, which could be controlled by Toll pathway.
Collapse
|
37
|
Lin CC, Chen YH, Guan TC, Chang SW, Pai H, Chou SJ, Tsai HP. Expression of foreign proteins by antimicrobial peptide gene promoters in mosquitoes. JOURNAL OF MEDICAL SCIENCES 2019. [DOI: 10.4103/jmedsci.jmedsci_194_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
38
|
Chen J, Zhang DW, Jin X, Xu XL, Zeng BP. Characterization of the Akirin Gene and Its Role in the NF-κB Signaling Pathway of Sogatella furcifera. Front Physiol 2018; 9:1411. [PMID: 30349487 PMCID: PMC6186838 DOI: 10.3389/fphys.2018.01411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/18/2018] [Indexed: 12/22/2022] Open
Abstract
Akirin is an essential nuclear protein involved in the regulation of NF-κB signaling pathway. In most invertebrates, Akirin regulates NF-κB-related Imd and Toll pathways, however, in Drosophila, it only controls the Imd pathway, whereas its role in NF-κB signaling pathway in other insect species is unclear. In the present study, we used white-backed planthopper Sogatella furcifera as a model to investigate the functional activity of Akirin in insects. The sequence of Akirin cDNA was extracted from transcriptome database of S. furcifera; it contained a 585 bp open reading frame (ORF) encoding a putative protein of 194 amino acids. S. furcifera Akirin (SfAkirin) had a molecular weight of about 21.69 kDa and a theoretical pI of 8.66 and included a nuclear localization signal (NLS) of five amino acid residues at the N-terminal region. Evolutionary analysis showed that SfAkirin was evolutionary closer to Akirins of such relatively distant species as crustaceans than to those of some insect orders like Diptera and Hymenoptera. Tissue-specific expression analysis showed that the SfAkirin gene was expressed in all examined tissues, with the highest expression levels detected in the testis, followed by the ovary, whereas the lowest expression was found in the head. Real-time quantitative PCR analysis showed that SfAkirin mRNA was strongly induced in response to injection of heat-inactivated Escherichia coli and Bacillus subtilis, whereas SfAkirin silencing by RNA interference significantly reduced the expression of NF-κB dependent transcription factors Dorsal and Relish after B. subtilis and E. coli challenge, respectively. Our results suggest that SfAkirin may control the immune response of S. furcifera against bacterial infection via both Imd and Toll signaling pathways.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| | - Dao-Wei Zhang
- School of Biological and Agricultural Science and Technology, Zunyi Normal University, Zunyi, China
| | - Xing Jin
- School of Biological and Agricultural Science and Technology, Zunyi Normal University, Zunyi, China
| | - Xian-Lin Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| | - Bo-Ping Zeng
- School of Biological and Agricultural Science and Technology, Zunyi Normal University, Zunyi, China
| |
Collapse
|
39
|
Shaw WR, Catteruccia F. Vector biology meets disease control: using basic research to fight vector-borne diseases. Nat Microbiol 2018; 4:20-34. [PMID: 30150735 DOI: 10.1038/s41564-018-0214-7] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
Abstract
Human pathogens that are transmitted by insects are a global problem, particularly those vectored by mosquitoes; for example, malaria parasites transmitted by Anopheles species, and viruses such as dengue, Zika and chikungunya that are carried by Aedes mosquitoes. Over the past 15 years, the prevalence of malaria has been substantially reduced and virus outbreaks have been contained by controlling mosquito vectors using insecticide-based approaches. However, disease control is now threatened by alarming rates of insecticide resistance in insect populations, prompting the need to develop a new generation of specific strategies that can reduce vector-mediated transmission. Here, we review how increased knowledge in insect biology and insect-pathogen interactions is stimulating new concepts and tools for vector control. We focus on strategies that either interfere with the development of pathogens within their vectors or directly impact insect survival, including enhancement of vector-mediated immune control, manipulation of the insect microbiome, or use of powerful new genetic tools such as CRISPR-Cas systems to edit vector genomes. Finally, we offer a perspective on the implementation hurdles as well as the knowledge gaps that must be filled in the coming years to safely realize the potential of these novel strategies to eliminate the scourge of vector-borne disease.
Collapse
Affiliation(s)
- W Robert Shaw
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| | - Flaminia Catteruccia
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| |
Collapse
|
40
|
Zhou J, Yu HY, Zhang W, Ahmad F, Hu SN, Zhao LL, Zou Z, Sun JH. Comparative analysis of the Monochamus alternatus immune system. INSECT SCIENCE 2018; 25:581-603. [PMID: 28247970 DOI: 10.1111/1744-7917.12453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/05/2017] [Accepted: 02/20/2017] [Indexed: 05/26/2023]
Abstract
The pine sawyer beetle, Monochamus alternatus, is regarded as a notorious forest pest in Asia, vectoring an invasive pathogenic nematode, Bursaphelenchus xylophilus, which is known to cause pine wilt disease. However, little sequence information is available for this vector beetle. This hampered the research on its immune system. Based on the transcriptome of M. alternatus, we have identified and characterized 194 immunity-related genes in M. alternatus, and compared them with homologues molecules from other species known to exhibit immune responses against invading microbes. The lower number of putative immunity-related genes in M. alternatus were attributed to fewer C-type lectin, serine protease (SP) and anti-microbial peptide (AMP) genes. Phylogenetic analysis revealed that M. alternatus had a unique recognition gene, galectin3, orthologues of which were not identified in Tribolium castaneum, Drosophila melanogastor, Anopheles gambiae and Apis mellifera. This suggested a lineage-specific gene evolution for coleopteran insects. Our study provides the comprehensive sequence resources of the immunity-related genes of M. alternatus, presenting valuable information for better understanding of the molecular mechanism of innate immunity processes in M. alternatus against B. xylophilus.
Collapse
Affiliation(s)
- Jiao Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hai-Ying Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Faheem Ahmad
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Song-Nian Hu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Li-Lin Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiang-Hua Sun
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
41
|
The C-Type Lectin Domain Gene Family in Aedes aegypti and Their Role in Arbovirus Infection. Viruses 2018; 10:v10070367. [PMID: 30002303 PMCID: PMC6070988 DOI: 10.3390/v10070367] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 12/15/2022] Open
Abstract
Several medically important flaviviruses that are transmitted by mosquitoes have been shown to bind to the C-type lectin fold that is present in either vertebrate or invertebrate proteins. While in some cases this interaction is part of a neutralizing anti-viral immune response, many reports have implicated this as critical for successful virus entry. Despite the establishment of mosquito C-type lectin domain containing proteins (CTLDcps) as known host factors in assisting the infectious process for flaviviruses, little is known about the structural characteristics of these proteins and their relationships to each other. In this report, we describe the manual annotation and structural characterization of 52 Aedes aegypti CTLDcps. Using existing RNAseq data, we establish that these genes can be subdivided into two classes: those highly conserved with expression primarily in development (embryo/early larvae) and those with no clear orthologs with expression primarily in late larvae/pupae or adults. The latter group contained all CTLDcps that are regulated by the Toll/Imd immune pathways, all known microbiome-regulating CTLDcps, and almost all CTLDcps that are implicated as flavivirus host factors in A. aegypti. Finally, we attempt to synthesize results from multiple conflicting gene expression profiling experiments in terms of how flavivirus infection changes steady-state levels of mRNA encoding CTLDcps.
Collapse
|
42
|
Serrato-Salas J, Izquierdo-Sánchez J, Argüello M, Conde R, Alvarado-Delgado A, Lanz-Mendoza H. Aedes aegypti antiviral adaptive response against DENV-2. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 84:28-36. [PMID: 29408269 DOI: 10.1016/j.dci.2018.01.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 06/07/2023]
Abstract
Priming is the conceptual term defining memory phenomenon in innate immune response. Numerous examples of enhanced secondary immune response have been described in diverse taxa of invertebrates; which naturally lacks memory response. In mosquitoes, a previous non-lethal challenge with some specific pathogens modify their immune response against the same microorganism; developing an improved antimicrobial reaction. In this work, we explore the ability of Aedes aegypti to mount a higher antiviral response upon a second oral DENV challenge. When previously challenged with inactive virus, we observed that the posterior infection showed a diminished number of DENV infectious particles in midguts and carcasses. In challenged tissues, we detected higher de novo midgut DNA synthesis than control group, as determined by DNA incorporation of 5-bromo-2-deoxyuridine. We demonstrated that inactive DENV particle are capable to induce DNA synthesis levels comparable to infective DENV. We considered the Drosophila melanogaster hindsight and Delta-Notch mosquitoes orthologues as potential de novo DNA synthesis pathway components (as observed in fly oocyte development and midgut tissue renewal). We showed that Aedes aegypti hindsight transcript relative expression levels were higher than control during DENV infection and inactive DENV particle alimentation. Also, Aedes aegypti second challenge with active DENV induced higher hindsight, Delta and Notch transcriptions in the primed mosquitoes (compared with the primary infection levels). Considering that the mosquito de novo DNA synthesis is concomitant to viral particle reduction, this finding opens a new perspective on the mechanisms underlying the vector antiviral immune response and the effector molecules involved.
Collapse
Affiliation(s)
- Javier Serrato-Salas
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico
| | - Javier Izquierdo-Sánchez
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico
| | - Martha Argüello
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico
| | - Renáud Conde
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico
| | - Alejandro Alvarado-Delgado
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico
| | - Humberto Lanz-Mendoza
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
43
|
Wang YH, Chang MM, Wang XL, Zheng AH, Zou Z. The immune strategies of mosquito Aedes aegypti against microbial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:12-21. [PMID: 29217264 DOI: 10.1016/j.dci.2017.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 06/07/2023]
Abstract
Yellow fever mosquito Aedes aegypti transmits many devastating arthropod-borne viruses (arboviruses), such as dengue virus, yellow fever virus, Chikungunya virus, and Zika virus, which cause great concern to human health. Mosquito control is an effective method to block the spread of infectious diseases. Ae. aegypti uses its innate immune system to fight against arboviruses, parasites, and fungi. In this review, we briefly summarize the recent findings in the immune response of Ae. aegypti against arboviral and entomopathogenic infections. This review enriches our understanding of the mosquito immune system and provides evidence to support the development of novel mosquito control strategies.
Collapse
Affiliation(s)
- Yan-Hong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng-Meng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Li Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ai-Hua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
44
|
Simões ML, Caragata EP, Dimopoulos G. Diverse Host and Restriction Factors Regulate Mosquito-Pathogen Interactions. Trends Parasitol 2018; 34:603-616. [PMID: 29793806 DOI: 10.1016/j.pt.2018.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Abstract
Mosquitoes transmit diseases that seriously impact global human health. Despite extensive knowledge of the life cycles of mosquito-borne parasites and viruses within their hosts, control strategies have proven insufficient to halt their spread. An understanding of the relationships established between such pathogens and the host tissues they inhabit is therefore paramount for the development of new strategies that specifically target these interactions, to prevent the pathogens' maturation and transmission. Here we present an updated account of the antagonists and host factors that affect the development of Plasmodium, the parasite causing malaria, and mosquito-borne viruses, such as dengue virus and Zika virus, within their mosquito vectors, and we discuss the similarities and differences between Plasmodium and viral systems, looking toward the elucidation of new targets for disease control.
Collapse
Affiliation(s)
- Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
45
|
Barrows NJ, Campos RK, Liao KC, Prasanth KR, Soto-Acosta R, Yeh SC, Schott-Lerner G, Pompon J, Sessions OM, Bradrick SS, Garcia-Blanco MA. Biochemistry and Molecular Biology of Flaviviruses. Chem Rev 2018; 118:4448-4482. [PMID: 29652486 DOI: 10.1021/acs.chemrev.7b00719] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Flaviviruses, such as dengue, Japanese encephalitis, tick-borne encephalitis, West Nile, yellow fever, and Zika viruses, are critically important human pathogens that sicken a staggeringly high number of humans every year. Most of these pathogens are transmitted by mosquitos, and not surprisingly, as the earth warms and human populations grow and move, their geographic reach is increasing. Flaviviruses are simple RNA-protein machines that carry out protein synthesis, genome replication, and virion packaging in close association with cellular lipid membranes. In this review, we examine the molecular biology of flaviviruses touching on the structure and function of viral components and how these interact with host factors. The latter are functionally divided into pro-viral and antiviral factors, both of which, not surprisingly, include many RNA binding proteins. In the interface between the virus and the hosts we highlight the role of a noncoding RNA produced by flaviviruses to impair antiviral host immune responses. Throughout the review, we highlight areas of intense investigation, or a need for it, and potential targets and tools to consider in the important battle against pathogenic flaviviruses.
Collapse
Affiliation(s)
- Nicholas J Barrows
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Department of Molecular Genetics and Microbiology , Duke University , Durham , North Carolina 27710 , United States
| | - Rafael K Campos
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Department of Molecular Genetics and Microbiology , Duke University , Durham , North Carolina 27710 , United States
| | - Kuo-Chieh Liao
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - K Reddisiva Prasanth
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Ruben Soto-Acosta
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Shih-Chia Yeh
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - Geraldine Schott-Lerner
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Julien Pompon
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore.,MIVEGEC, IRD, CNRS, Université de Montpellier , Montpellier 34090 , France
| | - October M Sessions
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| |
Collapse
|
46
|
Zhou J, Zhao LL, Yu HY, Wang YH, Zhang W, Hu SN, Zou Z, Sun JH. Immune tolerance of vector beetle to its partner plant parasitic nematode modulated by its insect parasitic nematode. FASEB J 2018; 32:4862-4877. [DOI: 10.1096/fj.201800247r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Jiao Zhou
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
| | - Li-Lin Zhao
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
| | - Hai-Ying Yu
- Key Laboratory of Genome Sciences and InformationBeijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Yan-Hong Wang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
| | - Wei Zhang
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
| | - Song-Nian Hu
- Key Laboratory of Genome Sciences and InformationBeijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jiang-Hua Sun
- State Key Laboratory of Integrated Management of Pest Insects and RodentsInstitute of ZoologyBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
47
|
Yeh SC, Pompon J. Flaviviruses Produce a Subgenomic Flaviviral RNA That Enhances Mosquito Transmission. DNA Cell Biol 2017; 37:154-159. [PMID: 29251994 DOI: 10.1089/dna.2017.4059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mosquito-borne flaviviruses (MBFVs) are a global public health burden. MBFVs have several unique 3'UTR structures that inhibit the host RNA decay machinery to produce subgenomic flaviviral RNAs (sfRNAs). Number of sfRNA species and their relative quantities are dependent on the 3'UTR tertiary structures and can vary between tissues. Two recent in vivo studies demonstrated that sfRNA enhances mosquito transmission, resulting in increased infection rate of saliva. Transmission efficiency is determined by the immune response. First evidence points to sfRNA interference with the Toll and RNAi immune pathways. However, a more complex picture that includes flexibility in sfRNA production and interaction with immune-related proteins remains to be explored.
Collapse
Affiliation(s)
- Shih-Chia Yeh
- 1 Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School , Singapore, Singapore
| | - Julien Pompon
- 1 Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School , Singapore, Singapore .,2 MIVEGEC, IRD, CNRS, University of Montpellier , Montpellier, France
| |
Collapse
|
48
|
Pan X, Pike A, Joshi D, Bian G, McFadden MJ, Lu P, Liang X, Zhang F, Raikhel AS, Xi Z. The bacterium Wolbachia exploits host innate immunity to establish a symbiotic relationship with the dengue vector mosquito Aedes aegypti. ISME JOURNAL 2017; 12:277-288. [PMID: 29099491 PMCID: PMC5739022 DOI: 10.1038/ismej.2017.174] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 01/15/2023]
Abstract
A host's immune system plays a central role in shaping the composition of the microbiota and, in return, resident microbes influence immune responses. Symbiotic associations of the maternally transmitted bacterium Wolbachia occur with a wide range of arthropods. It is, however, absent from the dengue and Zika vector mosquito Aedes aegypti in nature. When Wolbachia is artificially forced to form symbiosis with this new mosquito host, it boosts the basal immune response and enhances the mosquito's resistance to pathogens, including dengue, Zika virus and malaria parasites. The mechanisms involved in establishing a symbiotic relationship between Wolbachia and A. aegypti, and the long-term outcomes of this interaction, are not well understood. Here, we have demonstrated that both the immune deficiency (IMD) and Toll pathways are activated by the Wolbachia strain wAlbB upon its introduction into A. aegypti. Silencing the Toll and IMD pathways via RNA interference reduces the wAlbB load. Notably, wAlbB induces peptidoglycan recognition protein (PGRP)-LE expression in the carcass of A. aegypti, and its silencing results in a reduction of symbiont load. Using transgenic mosquitoes with stage-specific induction of the IMD and Toll pathways, we have shown that elevated wAlbB infection in these mosquitoes is maintained via maternal transmission. These results indicate that host innate immunity is utilized to establish and promote host-microbial symbiosis. Our results will facilitate a long-term projection of the stability of the Wolbachia-A. aegypti mosquito system that is being developed to control dengue and Zika virus transmission to humans.
Collapse
Affiliation(s)
- Xiaoling Pan
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,School of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Andrew Pike
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Deepak Joshi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Guowu Bian
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Michael J McFadden
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Peng Lu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Xiao Liang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Fengrui Zhang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Alexander S Raikhel
- Department of Entomology and Institute for Integrative Molecular Biology, University of California, Riverside, CA 92521, USA
| | - Zhiyong Xi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,Sun Yat-sen University-Michigan State University Joint Center of Vector Control for Tropical Diseases, Guangzhou, Guangdong 510080, China
| |
Collapse
|
49
|
Angleró-Rodríguez YI, MacLeod HJ, Kang S, Carlson JS, Jupatanakul N, Dimopoulos G. Aedes aegypti Molecular Responses to Zika Virus: Modulation of Infection by the Toll and Jak/Stat Immune Pathways and Virus Host Factors. Front Microbiol 2017; 8:2050. [PMID: 29109710 PMCID: PMC5660061 DOI: 10.3389/fmicb.2017.02050] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 10/06/2017] [Indexed: 12/23/2022] Open
Abstract
Zika (ZIKV) and dengue virus (DENV) are transmitted to humans by Aedes mosquitoes. However, the molecular interactions between the vector and ZIKV remain largely unexplored. In this work, we further investigated the tropism of ZIKV in two different Aedes aegypti strains and show that the virus infection kinetics, tissue migration, and susceptibility to infection differ between mosquito strains. We also compare the vector transcriptome changes upon ZIKV or DENV infection demonstrating that 40% of the mosquito's midgut infection-responsive transcriptome is virus-specific at 7 days after virus ingestion. Regulated genes included key factors of the mosquito's anti-viral immunity. Comparison of the ZIKV and DENV infection-responsive transcriptome data to those available for yellow fever virus and West Nile virus identified 26 genes likely to play key roles in virus infection of Aedes mosquitoes. Through reverse genetic analyses, we show that the Toll and the Jak/Stat innate immune pathways mediate increased resistance to ZIKV infection, and the conserved DENV host factors vATPase and inosine-5'-monophosphate dehydrogenase are also utilized for ZIKV infection.
Collapse
Affiliation(s)
- Yesseinia I Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Hannah J MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Jenny S Carlson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Natapong Jupatanakul
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
50
|
Pompon J, Manuel M, Ng GK, Wong B, Shan C, Manokaran G, Soto-Acosta R, Bradrick SS, Ooi EE, Missé D, Shi PY, Garcia-Blanco MA. Dengue subgenomic flaviviral RNA disrupts immunity in mosquito salivary glands to increase virus transmission. PLoS Pathog 2017; 13:e1006535. [PMID: 28753642 PMCID: PMC5555716 DOI: 10.1371/journal.ppat.1006535] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/14/2017] [Accepted: 07/15/2017] [Indexed: 12/21/2022] Open
Abstract
Globally re-emerging dengue viruses are transmitted from human-to-human by Aedes mosquitoes. While viral determinants of human pathogenicity have been defined, there is a lack of knowledge of how dengue viruses influence mosquito transmission. Identification of viral determinants of transmission can help identify isolates with high epidemiological potential. Additionally, mechanistic understanding of transmission will lead to better understanding of how dengue viruses harness evolution to cycle between the two hosts. Here, we identified viral determinants of transmission and characterized mechanisms that enhance production of infectious saliva by inhibiting immunity specifically in salivary glands. Combining oral infection of Aedes aegypti mosquitoes and reverse genetics, we identified two 3’ UTR substitutions in epidemic isolates that increased subgenomic flaviviral RNA (sfRNA) quantity, infectious particles in salivary glands and infection rate of saliva, which represents a measure of transmission. We also demonstrated that various 3’UTR modifications similarly affect sfRNA quantity in both whole mosquitoes and human cells, suggesting a shared determinism of sfRNA quantity. Furthermore, higher relative quantity of sfRNA in salivary glands compared to midgut and carcass pointed to sfRNA function in salivary glands. We showed that the Toll innate immune response was preferentially inhibited in salivary glands by viruses with the 3’UTR substitutions associated to high epidemiological fitness and high sfRNA quantity, pointing to a mechanism for higher saliva infection rate. By determining that sfRNA is an immune suppressor in a tissue relevant to mosquito transmission, we propose that 3’UTR/sfRNA sequence evolution shapes dengue epidemiology not only by influencing human pathogenicity but also by increasing mosquito transmission, thereby revealing a viral determinant of epidemiological fitness that is shared between the two hosts. Dengue is a re-emerging global disease transmitted from human-to-human by mosquitoes. While environmental and host immune factors are important, viral determinants of mosquito transmission also shape the epidemiology of dengue. Understanding how dengue viruses influence transmission will help identify isolates with high epidemic potential and untangle the evolutionary pressures at play in the dual-host cycle. Here, we identified 2 substitutions in the 3’UTR of epidemic isolates that increase transmission through immune suppression in the salivary glands. Using oral infection of Aedes aegypti mosquitoes, we reported that epidemic isolates produced more subgenomic flaviviral RNA (sfRNA) in salivary glands. SfRNA is generated from the 3’UTR sequence remaining after partial genome degradation by a host nuclease. Using reverse genetics, we identified the two 3’UTR substitutions responsible for the higher sfRNA quantity in salivary glands. We further showed that these substitutions increased dengue virus titer in salivary glands and rate of saliva infection, and suppressed the Toll immune response in salivary glands. Our study identifies the substitutions that determine virus epidemiological fitness and provides a mechanism for sfRNA-mediated enhancement of transmission. Together with previous work demonstrating that sfRNA sequence modification influences dengue virus pathogenicity in human, and that shows variation in sfRNA sequence when the viruses transition from one host to vector and vice versa, our study supports that sfRNA evolution is constrained in the two hosts.
Collapse
Affiliation(s)
- Julien Pompon
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- UMR IRD-CNRS MIVEGEC, IRD, Montpellier, France
- * E-mail: (JP); (MAGB)
| | - Menchie Manuel
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Geok Kee Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Benjamin Wong
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Gayathri Manokaran
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Ruben Soto-Acosta
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Shelton S. Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Mariano A. Garcia-Blanco
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
- * E-mail: (JP); (MAGB)
| |
Collapse
|