51
|
Short SM, Mongodin EF, MacLeod HJ, Talyuli OAC, Dimopoulos G. Amino acid metabolic signaling influences Aedes aegypti midgut microbiome variability. PLoS Negl Trop Dis 2017; 11:e0005677. [PMID: 28753661 PMCID: PMC5549995 DOI: 10.1371/journal.pntd.0005677] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 08/09/2017] [Accepted: 06/02/2017] [Indexed: 12/26/2022] Open
Abstract
The mosquito midgut microbiota has been shown to influence vector competence for multiple human pathogens. The microbiota is highly variable in the field, and the sources of this variability are not well understood, which limits our ability to understand or predict its effects on pathogen transmission. In this work, we report significant variation in female adult midgut bacterial load between strains of A. aegypti which vary in their susceptibility to dengue virus. Composition of the midgut microbiome was similar overall between the strains, with 81-92% of reads coming from the same five bacterial families, though we did detect differences in the presence of some bacterial families including Flavobacteriaceae and Entobacteriaceae. We conducted transcriptomic analysis on the two mosquito strains that showed the greatest difference in bacterial load, and found that they differ in transcript abundance of many genes implicated in amino acid metabolism, in particular the branched chain amino acid degradation pathway. We then silenced this pathway by targeting multiple genes using RNA interference, which resulted in strain-specific bacterial proliferation, thereby eliminating the difference in midgut bacterial load between the strains. This suggests that the branched chain amino acid (BCAA) degradation pathway controls midgut bacterial load, though the mechanism underlying this remains unclear. Overall, our results indicate that amino acid metabolism can act to influence the midgut microbiota. Moreover, they suggest that genetic or physiological variation in BCAA degradation pathway activity may in part explain midgut microbiota variation in the field.
Collapse
Affiliation(s)
- Sarah M. Short
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Emmanuel F. Mongodin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Hannah J. MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Octavio A. C. Talyuli
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 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
- * E-mail:
| |
Collapse
|
52
|
Domingos A, Pinheiro-Silva R, Couto J, do Rosário V, de la Fuente J. The Anopheles gambiae transcriptome - a turning point for malaria control. INSECT MOLECULAR BIOLOGY 2017; 26:140-151. [PMID: 28067439 DOI: 10.1111/imb.12289] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mosquitoes are important vectors of several pathogens and thereby contribute to the spread of diseases, with social, economic and public health impacts. Amongst the approximately 450 species of Anopheles, about 60 are recognized as vectors of human malaria, the most important parasitic disease. In Africa, Anopheles gambiae is the main malaria vector mosquito. Current malaria control strategies are largely focused on drugs and vector control measures such as insecticides and bed-nets. Improvement of current, and the development of new, mosquito-targeted malaria control methods rely on a better understanding of mosquito vector biology. An organism's transcriptome is a reflection of its physiological state and transcriptomic analyses of different conditions that are relevant to mosquito vector competence can therefore yield important information. Transcriptomic analyses have contributed significant information on processes such as blood-feeding parasite-vector interaction, insecticide resistance, and tissue- and stage-specific gene regulation, thereby facilitating the path towards the development of new malaria control methods. Here, we discuss the main applications of transcriptomic analyses in An. gambiae that have led to a better understanding of mosquito vector competence.
Collapse
Affiliation(s)
- A Domingos
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
- Global Health and Tropical Medicine (GHMT), Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - R Pinheiro-Silva
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - J Couto
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - V do Rosário
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - J de la Fuente
- SaBio. Instituto de Investigación de Recursos Cinegéticos, IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
53
|
Hormone and receptor interplay in the regulation of mosquito lipid metabolism. Proc Natl Acad Sci U S A 2017; 114:E2709-E2718. [PMID: 28292900 DOI: 10.1073/pnas.1619326114] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquitoes transmit devastating human diseases because they need vertebrate blood for egg development. Metabolism in female mosquitoes is tightly coupled with blood meal-mediated reproduction, which requires an extremely high level of energy consumption. Functional analysis has shown that major genes encoding for enzymes involved in lipid metabolism (LM) in the mosquito fat bodies are down-regulated at the end of the juvenile hormone (JH)-controlled posteclosion (PE) phase but exhibit significant elevation in their transcript levels during the post-blood meal phase (PBM), which is regulated mainly by 20-hydroxyecdysone (20E). Reductions in the transcript levels of genes encoding triacylglycerol (TAG) catabolism and β-oxidation enzymes were observed to correlate with a dramatic accumulation of lipids in the PE phase; in contrast, these transcripts were elevated significantly and lipid stores were diminished during the PBM phase. The RNAi depletion of Methoprene-tolerant (Met) and ecdysone receptor (EcR), receptors for JH and 20E, respectively, reversed the LM gene expression and the levels of lipid stores and metabolites, demonstrating the critical roles of these hormones in LM regulation. Hepatocyte nuclear factor 4 (HNF4) RNAi-silenced mosquitoes exhibited down-regulation of the gene transcripts encoding TAG catabolism and β-oxidation enzymes and an inability to use lipids effectively, as manifested by TAG accumulation. The luciferase reporter assay showed direct regulation of LM-related genes by HNF4. Moreover, HNF4 gene expression was down-regulated by Met and activated by EcR and Target of rapamycin, providing a link between nutritional and hormonal regulation of LM in female mosquitoes.
Collapse
|
54
|
Barletta ABF, Nascimento-Silva MCL, Talyuli OAC, Oliveira JHM, Pereira LOR, Oliveira PL, Sorgine MHF. Microbiota activates IMD pathway and limits Sindbis infection in Aedes aegypti. Parasit Vectors 2017; 10:103. [PMID: 28231846 PMCID: PMC5324288 DOI: 10.1186/s13071-017-2040-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 02/16/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Aedes aegypti is the main vector of important arboviruses such as dengue, Zika and chikungunya. During infections mosquitoes can activate the immune pathways Toll, IMD and JAK/STAT to limit pathogen replication. RESULTS Here, we evaluate the immune response profile of Ae. aegypti against Sindbis virus (SINV). We analyzed gene expression of components of Toll, IMD and JAK/STAT pathways and showed that a blood meal and virus infection upregulated aaREL2 in a microbiota-dependent fashion, since this induction was prevented by antibiotic. The presence of the microbiota activates IMD and impaired the replication of SINV in the midgut. Constitutive activation of the IMD pathway, by Caspar depletion, leads to a decrease in microbiota levels and an increase in SINV loads. CONCLUSION Together, these results suggest that a blood meal is able to activate innate immune pathways, through a nutrient induced growth of microbiota, leading to upregulation of aaREL2 and IMD activation. Microbiota levels seemed to have a reciprocal interaction, where the proliferation of the microbiota activates IMD pathway that in turn controls bacterial levels, allowing SINV replication in Ae. aegypti mosquitoes. The activation of the IMD pathway seems to have an indirect effect in SINV levels that is induced by the microbiota.
Collapse
Affiliation(s)
- Ana Beatriz Ferreira Barletta
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | - Octávio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Henrique M Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Henrique F Sorgine
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
55
|
Jupatanakul N, Sim S, Angleró-Rodríguez YI, Souza-Neto J, Das S, Poti KE, Rossi SL, Bergren N, Vasilakis N, Dimopoulos G. Engineered Aedes aegypti JAK/STAT Pathway-Mediated Immunity to Dengue Virus. PLoS Negl Trop Dis 2017; 11:e0005187. [PMID: 28081143 PMCID: PMC5230736 DOI: 10.1371/journal.pntd.0005187] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/15/2016] [Indexed: 12/31/2022] Open
Abstract
We have developed genetically modified Ae. aegypti mosquitoes that activate the conserved antiviral JAK/STAT pathway in the fat body tissue, by overexpressing either the receptor Dome or the Janus kinase Hop by the blood feeding-induced vitellogenin (Vg) promoter. Transgene expression inhibits infection with several dengue virus (DENV) serotypes in the midgut as well as systemically and in the salivary glands. The impact of the transgenes Dome and Hop on mosquito longevity was minimal, but it resulted in a compromised fecundity when compared to wild-type mosquitoes. Overexpression of Dome and Hop resulted in profound transcriptome regulation in the fat body tissue as well as the midgut tissue, pinpointing several expression signatures that reflect mechanisms of DENV restriction. Our transcriptome studies and reverse genetic analyses suggested that enrichment of DENV restriction factor and depletion of DENV host factor transcripts likely accounts for the DENV inhibition, and they allowed us to identify novel factors that modulate infection. Interestingly, the fat body-specific activation of the JAK/STAT pathway did not result in any enhanced resistance to Zika virus (ZIKV) or chikungunya virus (CHIKV) infection, thereby indicating a possible specialization of the pathway’s antiviral role. Dengue has represented a significant public health burden for a number of decades, and given the lack of dengue-specific drugs and limited availability of licensed vaccine, new methods for prevention and control are urgently needed. Here, we investigated whether genetic manipulation of the mosquitoes’ native JAK/STAT pathway-mediated anti-DENV defense system could be used to render mosquitoes more resistant to infection. We generated Ae. aegypti mosquitoes overexpressing the JAK/STAT pathway components Dome and Hop under the control of a bloodmeal-inducible, fat body-specific vitellogenin (Vg) promoter. These genetically modified mosquitoes showed an increased resistance to DENV infection, likely because of higher expression of DENV restriction factors and lower expression of DENV host factors, as indicated by transcriptome analyses. Expression of the transgenes had a minimal impact on mosquito longevity; however, it significantly impaired the mosquitoes’ fecundity. Interestingly, bloodmeal-inducible fat body-specific overexpression of either Hop or Dome did not affect mosquito permissiveness to either ZIKV or CHIKV infection, suggesting a possible specialization of JAK/STAT pathway antiviral defenses. Thus, our study is the first to provide a proof-of-concept that genetic engineering of the mosquitoes’ JAK/STAT immune pathway can be used to render this host more resistant to DENV infection.
Collapse
Affiliation(s)
- Natapong Jupatanakul
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shuzhen Sim
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yesseinia I. Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jayme Souza-Neto
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Suchismita Das
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kristin E. Poti
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shannan L. Rossi
- Department of Pathology and Center of Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston TX, United States of America
| | - Nicholas Bergren
- Department of Pathology and Center of Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston TX, United States of America
| | - Nikos Vasilakis
- Department of Pathology and Center of Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston TX, 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
- * E-mail:
| |
Collapse
|
56
|
Xu J, Morisseau C, Yang J, Lee KSS, Kamita SG, Hammock BD. Ingestion of the epoxide hydrolase inhibitor AUDA modulates immune responses of the mosquito, Culex quinquefasciatus during blood feeding. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 76:62-69. [PMID: 27369469 PMCID: PMC5010450 DOI: 10.1016/j.ibmb.2016.06.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 05/14/2023]
Abstract
Epoxide hydrolases (EHs) are enzymes that play roles in metabolizing xenobiotic epoxides from the environment, and in regulating lipid signaling molecules, such as juvenile hormones in insects and epoxy fatty acids in mammals. In this study we fed mosquitoes with an epoxide hydrolase inhibitor AUDA during artificial blood feeding, and we found the inhibitor increased the concentration of epoxy fatty acids in the midgut of female mosquitoes. We also observed ingestion of AUDA triggered early expression of defensin A, cecropin A and cecropin B2 at 6 h after blood feeding. The expression of cecropin B1 and gambicin were not changed more than two fold compared to controls. The changes in gene expression were transient possibly because more than 99% of the inhibitor was metabolized or excreted at 42 h after being ingested. The ingestion of AUDA also affected the growth of bacteria colonizing in the midgut, but did not affect mosquito longevity, fecundity and fertility in our laboratory conditions. When spiked into the blood, EpOMEs and DiHOMEs were as effective as the inhibitor AUDA in reducing the bacterial load in the midgut, while EETs rescued the effects of AUDA. Our data suggest that epoxy fatty acids from host blood are immune response regulators metabolized by epoxide hydrolases in the midgut of female mosquitoes, inhibition of which causes transient changes in immune responses, and affects growth of microbes in the midgut.
Collapse
Affiliation(s)
- Jiawen Xu
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Christophe Morisseau
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Jun Yang
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Kin Sing Stephen Lee
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Shizuo G Kamita
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Bruce D Hammock
- Department of Entomology and UC Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA.
| |
Collapse
|
57
|
Liu YX, Li FX, Liu ZZ, Jia ZR, Zhou YH, Zhang H, Yan H, Zhou XQ, Chen XG. Integrated analysis of miRNAs and transcriptomes in Aedes albopictus midgut reveals the differential expression profiles of immune-related genes during dengue virus serotype-2 infection. INSECT SCIENCE 2016; 23:377-385. [PMID: 27029517 DOI: 10.1111/1744-7917.12339] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 03/12/2016] [Accepted: 03/21/2016] [Indexed: 06/05/2023]
Abstract
Mosquito microRNAs (miRNAs) are involved in host-virus interaction, and have been reported to be altered by dengue virus (DENV) infection in Aedes albopictus (Diptera: Culicidae). However, little is known about the molecular mechanisms of Aedes albopictus midgut-the first organ to interact with DENV-involved in its resistance to DENV. Here we used high-throughput sequencing to characterize miRNA and messenger RNA (mRNA) expression patterns in Aedes albopictus midgut in response to dengue virus serotype 2. A total of three miRNAs and 777 mRNAs were identified to be differentially expressed upon DENV infection. For the mRNAs, we identified 198 immune-related genes and 31 of them were differentially expressed. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses also showed that the differentially expressed immune-related genes were involved in immune response. Then the differential expression patterns of six immune-related genes and three miRNAs were confirmed by real-time reverse transcription polymerase chain reaction. Furthermore, seven known miRNA-mRNA interaction pairs were identified by aligning our two datasets. These analyses of miRNA and mRNA transcriptomes provide valuable information for uncovering the DENV response genes and provide a basis for future study of the resistance mechanisms in Aedes albopictus midgut.
Collapse
Affiliation(s)
- Yan-Xia Liu
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
- Laboratory of Emerging Infectious Diseases and Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fen-Xiang Li
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Zhuan-Zhuan Liu
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Zhi-Rong Jia
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Yan-He Zhou
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hao Zhang
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hui Yan
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | | | - Xiao-Guang Chen
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
58
|
Peuß R, Wensing KU, Woestmann L, Eggert H, Milutinović B, Sroka MGU, Scharsack JP, Kurtz J, Armitage SAO. Down syndrome cell adhesion molecule 1: testing for a role in insect immunity, behaviour and reproduction. ROYAL SOCIETY OPEN SCIENCE 2016; 3:160138. [PMID: 27152227 PMCID: PMC4852650 DOI: 10.1098/rsos.160138] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/22/2016] [Indexed: 05/24/2023]
Abstract
Down syndrome cell adhesion molecule 1 (Dscam1) has wide-reaching and vital neuronal functions although the role it plays in insect and crustacean immunity is less well understood. In this study, we combine different approaches to understand the roles that Dscam1 plays in fitness-related contexts in two model insect species. Contrary to our expectations, we found no short-term modulation of Dscam1 gene expression after haemocoelic or oral bacterial exposure in Tribolium castaneum, or after haemocoelic bacterial exposure in Drosophila melanogaster. Furthermore, RNAi-mediated Dscam1 knockdown and subsequent bacterial exposure did not reduce T. castaneum survival. However, Dscam1 knockdown in larvae resulted in adult locomotion defects, as well as dramatically reduced fecundity in males and females. We suggest that Dscam1 does not always play a straightforward role in immunity, but strongly influences behaviour and fecundity. This study takes a step towards understanding more about the role of this intriguing gene from different phenotypic perspectives.
Collapse
|
59
|
Cheng G, Liu Y, Wang P, Xiao X. Mosquito Defense Strategies against Viral Infection. Trends Parasitol 2016; 32:177-186. [PMID: 26626596 PMCID: PMC4767563 DOI: 10.1016/j.pt.2015.09.009] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 01/23/2023]
Abstract
Mosquito-borne viral diseases are a major concern of global health and result in significant economic losses in many countries. As natural vectors, mosquitoes are very permissive to and allow systemic and persistent arbovirus infection. Intriguingly, persistent viral propagation in mosquito tissues neither results in dramatic pathological sequelae nor impairs the vectorial behavior or lifespan, indicating that mosquitoes have evolved mechanisms to tolerate persistent infection and developed efficient antiviral strategies to restrict viral replication to nonpathogenic levels. Here we provide an overview of recent progress in understanding mosquito antiviral immunity and advances in the strategies by which mosquitoes control viral infection in specific tissues.
Collapse
Affiliation(s)
- Gong Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China.
| | - Yang Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China; School of Life Science, Tsinghua University, Beijing 100084, PR China
| | - Penghua Wang
- Department of Microbiology and Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595, USA
| | - Xiaoping Xiao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China
| |
Collapse
|
60
|
Jain S, Shrinet J, Tridibes A, Bhatnagar RK, Sunil S. miRNA⁻mRNA Conflux Regulating Immunity and Oxidative Stress Pathways in the Midgut of Blood-Fed Anopheles stephensi. Noncoding RNA 2015; 1:222-245. [PMID: 29861425 PMCID: PMC5932549 DOI: 10.3390/ncrna1030222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/02/2015] [Accepted: 11/11/2015] [Indexed: 12/13/2022] Open
Abstract
Blood feeding in Anopheles stephensi initiates a cascade of events that modulate several physiological functions in the mosquito. The midgut epithelium activates several of its molecules, most important among these being microRNAs, which regulate some of the physiological changes by targeting diverse mRNAs. The present study was conducted to identify and evaluate interactions between targets of eight miRNAs that are regulated on blood feeding. Identified from our previous study, we show these eight miRNAs exhibited distinct tissue specific expression. Targets of these miRNAs were predicted using computational approaches involving bioinformatics, co-expression analysis of the transcriptome and miRNome of blood-fed An. stephensi midgut. Using degradome sequencing, we identified some cleaved mRNAs of these microRNAs and, by using antagomiR knockdown technology to repress the miRNAs, the targets were validated in an An. stephensi cell line and in An. stephensi mosquitoes. In-depth analysis of predicted and identified targets revealed that the regulated miRNAs modulate well-characterized molecules that are involved in combating oxidative stress and immunity pathways through a dynamic miRNA:mRNA network. Our study is the first to identify miRNA:mRNA interactomes that play important role in maintaining redox homeostasis during blood feeding in the midgut of An. stephensi.
Collapse
Affiliation(s)
- Shanu Jain
- Insect Resistance Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| | - Jatin Shrinet
- Insect Resistance Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| | - Adak Tridibes
- National Institute of Malaria Research, Dwarka, New Delhi-110077, India.
| | - Raj K Bhatnagar
- Insect Resistance Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| | - Sujatha Sunil
- Insect Resistance Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| |
Collapse
|
61
|
Kvist J, Mattila ALK, Somervuo P, Ahola V, Koskinen P, Paulin L, Salmela L, Fountain T, Rastas P, Ruokolainen A, Taipale M, Holm L, Auvinen P, Lehtonen R, Frilander MJ, Hanski I. Flight-induced changes in gene expression in the Glanville fritillary butterfly. Mol Ecol 2015; 24:4886-900. [DOI: 10.1111/mec.13359] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Jouni Kvist
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 9) Helsinki Finland
| | - Anniina L. K. Mattila
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Panu Somervuo
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 9) Helsinki Finland
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 4) Helsinki Finland
| | - Virpi Ahola
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Patrik Koskinen
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 9) Helsinki Finland
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 4) Helsinki Finland
| | - Lars Paulin
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 4) Helsinki Finland
| | - Leena Salmela
- Department of Computer Science and Helsinki Institute for Information Technology HIIT; University of Helsinki; P.O. Box 68 (Gustaf Hällströmin katu 2b) Helsinki Finland
| | - Toby Fountain
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Pasi Rastas
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Annukka Ruokolainen
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Minna Taipale
- Science for Life Laboratory; Department of Biosciences and Nutrition; Karolinska Institutet (Hälsovägen 7); SE-14157 Huddinge Sweden
| | - Liisa Holm
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 4) Helsinki Finland
| | - Petri Auvinen
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 4) Helsinki Finland
| | - Rainer Lehtonen
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| | - Mikko J. Frilander
- Institute of Biotechnology; University of Helsinki; P.O. Box 56 (Viikinkaari 9) Helsinki Finland
| | - Ilkka Hanski
- Department of Biosciences; University of Helsinki; P.O. Box 65 (Viikinkaari 1) Helsinki FI-00014 Finland
| |
Collapse
|
62
|
Paradkar PN, Duchemin JB, Rodriguez-Andres J, Trinidad L, Walker PJ. Cullin4 Is Pro-Viral during West Nile Virus Infection of Culex Mosquitoes. PLoS Pathog 2015; 11:e1005143. [PMID: 26325027 PMCID: PMC4556628 DOI: 10.1371/journal.ppat.1005143] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023] Open
Abstract
Although mosquitoes serve as vectors of many pathogens of public health importance, their response to viral infection is poorly understood. It also remains to be investigated whether viruses deploy some mechanism to be able to overcome this immune response. Here, we have used an RNA-Seq approach to identify differentially regulated genes in Culex quinquefasciatus cells following West Nile virus (WNV) infection, identifying 265 transcripts from various cellular pathways that were either upregulated or downregulated. Ubiquitin-proteasomal pathway genes, comprising 12% of total differentially regulated genes, were selected for further validation by real time RT-qPCR and functional analysis. It was found that treatment of infected cells with proteasomal inhibitor, MG-132, decreased WNV titers, indicating importance of this pathway during infection process. In infection models, the Culex ortholog of mammalian Cul4A/B (cullin RING ubiquitin ligase) was found to be upregulated in vitro as well as in vivo, especially in midguts of mosquitoes. Gene knockdown using dsRNA and overexpression studies indicated that Culex Cul4 acts as a pro-viral protein by degradation of CxSTAT via ubiquitin-proteasomal pathway. We also show that gene knockdown of Culex Cul4 leads to activation of the Jak-STAT pathway in mosquitoes leading to decrease viral replication in the body as well as saliva. Our results suggest a novel mechanism adopted by WNV to overcome mosquito immune response and increase viral replication. Mosquitoes are responsible for transmitting a large number of human and livestock viruses, like West Nile, dengue and Japanese encephalitis viruses. Infection of female mosquitoes with these viruses during blood feeding elicits an immune response. It is not known how the viruses manage to replicate in spite of this antiviral response. We used an unbiased transcriptome sequencing approach to identify genes differentially regulated after WNV infection resulting in 265 transcripts from various cellular pathways. Ubiquitin-proteasomal pathway, responsible for protein degradation, was found to be important during viral infection in mosquito cells. Using in vitro and in vivo infection models, we identified Culex Cul4 to be acting as pro-viral protein, increasing viral titers. Knockdown of Cul4 in Culex mosquitoes decreased viral titers in mosquito saliva. Identification of this novel immune evasion mechanism adopted by WNV provides new insights into transmission of arbovirus and interaction of WNV with its mosquito vector.
Collapse
Affiliation(s)
- Prasad N. Paradkar
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
- * E-mail:
| | - Jean-Bernard Duchemin
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Julio Rodriguez-Andres
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Lee Trinidad
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Peter J. Walker
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| |
Collapse
|
63
|
Roy S, Saha TT, Johnson L, Zhao B, Ha J, White KP, Girke T, Zou Z, Raikhel AS. Regulation of Gene Expression Patterns in Mosquito Reproduction. PLoS Genet 2015; 11:e1005450. [PMID: 26274815 PMCID: PMC4537244 DOI: 10.1371/journal.pgen.1005450] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/17/2015] [Indexed: 12/14/2022] Open
Abstract
In multicellular organisms, development, growth and reproduction require coordinated expression of numerous functional and regulatory genes. Insects, in addition to being the most speciose animal group with enormous biological and economical significance, represent outstanding model organisms for studying regulation of synchronized gene expression due to their rapid development and reproduction. Disease-transmitting female mosquitoes have adapted uniquely for ingestion and utilization of the huge blood meal required for swift reproductive events to complete egg development within a 72-h period. We investigated the network of regulatory factors mediating sequential gene expression in the fat body, a multifunctional organ analogous to the vertebrate liver and adipose tissue, of the female Aedes aegypti mosquito. Transcriptomic and bioinformatics analyses revealed that ~7500 transcripts are differentially expressed in four sequential waves during the 72-h reproductive period. A combination of RNA-interference gene-silencing and in-vitro organ culture identified the major regulators for each of these waves. Amino acids (AAs) regulate the first wave of gene activation between 3 h and 12 h post-blood meal (PBM). During the second wave, between 12 h and 36 h, most genes are highly upregulated by a synergistic action of AAs, 20-hydroxyecdysone (20E) and the Ecdysone-Receptor (EcR). Between 36 h and 48 h, the third wave of gene activation—regulated mainly by HR3—occurs. Juvenile Hormone (JH) and its receptor Methoprene-Tolerant (Met) are major regulators for the final wave between 48 h and 72 h. Each of these key regulators also has repressive effects on one or more gene sets. Our study provides a better understanding of the complexity of the regulatory mechanisms related to temporal coordination of gene expression during reproduction. We have detected the novel function of 20E/EcR responsible for transcriptional repression. This study also reveals the previously unidentified large-scale effects of HR3 and JH/Met on transcriptional regulation during the termination of vitellogenesis and remodeling of the fat body. In addition to being vectors of devastating human diseases, mosquitoes represent outstanding model organisms for studying regulatory mechanisms of differential gene expression due to their rapid reproductive cycles. About 7500 transcripts are differentially expressed in four sequential waves during the 72-h reproductive period in the fat body, a critical reproductive organ. The major regulators for these waves of gene expression are the two very important insect hormones, 20-hydroxyecdysone (20E) and Juvenile hormone (JH), their respective receptors Ecdysone Receptor (EcR) and Methoprene-Tolerant (Met), amino acids and the orphan nuclear receptor HR3. These key regulators are responsible for activation and repression of co-regulated gene sets, at different time points, within the 72-h reproductive period. Importantly, this study, apart from providing an insight into the regulatory complexity involved in the temporal coordination of gene expression, also reveals the previously unidentified roles of 20E/EcR, JH/Met and HR3 during the 72-h period post blood meal.
Collapse
Affiliation(s)
- Sourav Roy
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Tusar T. Saha
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Lisa Johnson
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Graduate Program in Cell, Molecular and Developmental Biology, University of California, Riverside, Riverside, California, United States of America
| | - Bo Zhao
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
| | - Jisu Ha
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Graduate Program in Genetics, Genomics and Bioinformatics, University of California, Riverside, Riverside, California, United States of America
| | - Kevin P. White
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, Illinois, United States of America
| | - Thomas Girke
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California, United States of America
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- * E-mail: (ZZ); (ASR)
| | - Alexander S. Raikhel
- Department of Entomology, University of California, Riverside, Riverside, California, United States of America
- Institute of Integrative Genome Biology, University of California, Riverside, Riverside, California, United States of America
- * E-mail: (ZZ); (ASR)
| |
Collapse
|
64
|
Wang YH, Hu Y, Xing LS, Jiang H, Hu SN, Raikhel AS, Zou Z. A Critical Role for CLSP2 in the Modulation of Antifungal Immune Response in Mosquitoes. PLoS Pathog 2015; 11:e1004931. [PMID: 26057557 PMCID: PMC4461313 DOI: 10.1371/journal.ppat.1004931] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 05/04/2015] [Indexed: 11/18/2022] Open
Abstract
Entomopathogenic fungi represent a promising class of bio-insecticides for mosquito control. Thus, detailed knowledge of the molecular mechanisms governing anti-fungal immune response in mosquitoes is essential. In this study, we show that CLSP2 is a modulator of immune responses during anti-fungal infection in the mosquito Aedes aegypti. With a fungal infection, the expression of the CLSP2 gene is elevated. CLSP2 is cleaved upon challenge with Beauveria bassiana conidia, and the liberated CLSP2 CTL-type domain binds to fungal cell components and B. bassiana conidia. Furthermore, CLPS2 RNA interference silencing significantly increases the resistance to the fungal challenge. RNA-sequencing transcriptome analysis showed that the majority of immune genes were highly upregulated in the CLSP2-depleted mosquitoes infected with the fungus. The up-regulated immune gene cohorts belong to melanization and Toll pathways, but not to the IMD or JAK-STAT. A thioester-containing protein (TEP22), a member of α2-macroglobulin family, has been implicated in the CLSP2-modulated mosquito antifungal defense. Our study has contributed to a greater understanding of immune-modulating mechanisms in mosquitoes.
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, China
| | - Yang Hu
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Long-Sheng Xing
- 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
| | - Hong Jiang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Song-Nian Hu
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Alexander S. Raikhel
- Department of Entomology and Institute for Integrative Genome Biology, University of California, Riverside, 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, China
- * E-mail: (ASR); (ZZ)
| |
Collapse
|
65
|
Juneja P, Ariani CV, Ho YS, Akorli J, Palmer WJ, Pain A, Jiggins FM. Exome and transcriptome sequencing of Aedes aegypti identifies a locus that confers resistance to Brugia malayi and alters the immune response. PLoS Pathog 2015; 11:e1004765. [PMID: 25815506 PMCID: PMC4376896 DOI: 10.1371/journal.ppat.1004765] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 02/25/2015] [Indexed: 11/18/2022] Open
Abstract
Many mosquito species are naturally polymorphic for their abilities to transmit parasites, a feature which is of great interest for controlling vector-borne disease. Aedes aegypti, the primary vector of dengue and yellow fever and a laboratory model for studying lymphatic filariasis, is genetically variable for its capacity to harbor the filarial nematode Brugia malayi. The genome of Ae. aegypti is large and repetitive, making genome resequencing difficult and expensive. We designed exome captures to target protein-coding regions of the genome, and used association mapping in a wild Kenyan population to identify a single, dominant, sex-linked locus underlying resistance. This falls in a region of the genome where a resistance locus was previously mapped in a line established in 1936, suggesting that this polymorphism has been maintained in the wild for the at least 80 years. We then crossed resistant and susceptible mosquitoes to place both alleles of the gene into a common genetic background, and used RNA-seq to measure the effect of this locus on gene expression. We found evidence for Toll, IMD, and JAK-STAT pathway activity in response to early stages of B. malayi infection when the parasites are beginning to die in the resistant genotype. We also found that resistant mosquitoes express anti-microbial peptides at the time of parasite-killing, and that this expression is suppressed in susceptible mosquitoes. Together, we have found that a single resistance locus leads to a higher immune response in resistant mosquitoes, and we identify genes in this region that may be responsible for this trait.
Collapse
Affiliation(s)
- Punita Juneja
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Cristina V. Ariani
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Yung Shwen Ho
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | - Jewelna Akorli
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, Accra, Ghana
| | - William J. Palmer
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Arnab Pain
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | - Francis M. Jiggins
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
66
|
Armitage SAO, Peuss R, Kurtz J. Dscam and pancrustacean immune memory - a review of the evidence. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 48:315-323. [PMID: 24657209 DOI: 10.1016/j.dci.2014.03.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/04/2014] [Accepted: 03/09/2014] [Indexed: 06/03/2023]
Abstract
Evidence is accumulating for a memory-like phenomenon in the immune defence of invertebrates. Down syndrome cell adhesion molecule (Dscam) has been proposed as a key candidate for a somatically diversified receptor system in the crustaceans and insects (Pancrustacea) that could enable challenge-specific protection. However, what is the evidence for an involvement of Dscam in pancrustacean immune memory, and in particular specificity? Here we review the current state of the art, and discuss hypotheses of how Dscam could be involved in immunity. We conclude that while there is increasing evidence for the involvement of Dscam in pancrustacean immunity, crucial experiments to address whether it plays a role in specificity upon secondary encounter with a pathogen still remain to be done.
Collapse
Affiliation(s)
- Sophie A O Armitage
- Institute for Evolution and Biodiversity, University of Münster, Hüfferstrasse 1, 48149 Münster, Germany.
| | - Robert Peuss
- 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
|
67
|
Yu J, Sun Q, Tang M. The nonlinear dynamics and fluctuations of mRNA levels in cross-talking pathway activated transcription. J Theor Biol 2014; 363:223-34. [DOI: 10.1016/j.jtbi.2014.08.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 08/12/2014] [Accepted: 08/13/2014] [Indexed: 11/29/2022]
|
68
|
Pike A, Vadlamani A, Sandiford SL, Gacita A, Dimopoulos G. Characterization of the Rel2-regulated transcriptome and proteome of Anopheles stephensi identifies new anti-Plasmodium factors. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 52:82-93. [PMID: 24998399 PMCID: PMC4143444 DOI: 10.1016/j.ibmb.2014.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/22/2014] [Accepted: 06/16/2014] [Indexed: 06/03/2023]
Abstract
Mosquitoes possess an innate immune system that is capable of limiting infection by a variety of pathogens, including the Plasmodium spp. parasites responsible for human malaria. The Anopheles immune deficiency (IMD) innate immune signaling pathway confers resistance to Plasmodium falciparum. While some previously identified Anopheles anti-Plasmodium effectors are regulated through signaling by Rel2, the transcription factor of the IMD pathway, many components of this defense system remain uncharacterized. To begin to better understand the regulation of immune effector proteins by the IMD pathway, we used oligonucleotide microarrays and iTRAQ to analyze differences in mRNA and protein expression, respectively, between transgenic Anopheles stephensi mosquitoes exhibiting blood meal-inducible overexpression of an active recombinant Rel2 and their wild-type conspecifics. Numerous genes were differentially regulated at both the mRNA and protein levels following induction of Rel2. While multiple immune genes were up-regulated, a majority of the differentially expressed genes have no known immune function in mosquitoes. Selected up-regulated genes from multiple functional categories were tested for both anti-Plasmodium and anti-bacterial action using RNA interference (RNAi). Based on our experimental findings, we conclude that increased expression of the IMD immune pathway-controlled transcription factor Rel2 affects the expression of numerous genes with diverse functions, suggesting a broader physiological impact of immune activation and possible functional versatility of Rel2. Our study has also identified multiple novel genes implicated in anti-Plasmodium defense.
Collapse
Affiliation(s)
- Andrew Pike
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Alekhya Vadlamani
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Simone L Sandiford
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Anthony Gacita
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| |
Collapse
|
69
|
Rückert C, Bell-Sakyi L, Fazakerley JK, Fragkoudis R. Antiviral responses of arthropod vectors: an update on recent advances. Virusdisease 2014; 25:249-60. [PMID: 25674592 DOI: 10.1007/s13337-014-0217-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 04/30/2014] [Indexed: 01/24/2023] Open
Abstract
Arthropod vectors, such as mosquitoes, ticks, biting midges and sand flies, transmit many viruses that can cause outbreaks of disease in humans and animals around the world. Arthropod vector species are invading new areas due to globalisation and environmental changes, and contact between exotic animal species, humans and arthropod vectors is increasing, bringing with it the regular emergence of new arboviruses. For future strategies to control arbovirus transmission, it is important to improve our understanding of virus-vector interactions. In the last decade knowledge of arthropod antiviral immunity has increased rapidly. RNAi has been proposed as the most important antiviral response in mosquitoes and it is likely to be the most important antiviral response in all arthropods. However, other newly-discovered antiviral strategies such as melanisation and the link between RNAi and the JAK/STAT pathway via the cytokine Vago have been characterised in the last few years. This review aims to summarise the most important and most recent advances made in arthropod antiviral immunity.
Collapse
Affiliation(s)
- Claudia Rückert
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF UK ; The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | | | | | | |
Collapse
|
70
|
Paradkar PN, Duchemin JB, Voysey R, Walker PJ. Dicer-2-dependent activation of Culex Vago occurs via the TRAF-Rel2 signaling pathway. PLoS Negl Trop Dis 2014; 8:e2823. [PMID: 24762775 PMCID: PMC3998923 DOI: 10.1371/journal.pntd.0002823] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/11/2014] [Indexed: 01/17/2023] Open
Abstract
Despite their importance as vectors of human and livestock diseases, relatively little is known about innate antiviral immune pathways in mosquitoes and other insects. Previous work has shown that Culex Vago (CxVago), which is induced and secreted from West Nile virus (WNV)-infected mosquito cells, acts as a functional homolog of interferon, by activating Jak-STAT pathway and limiting virus replication in neighbouring cells. Here we describe the Dicer-2-dependent pathway leading to WNV-induced CxVago activation. Using a luciferase reporter assay, we show that a NF-κB-like binding site in CxVago promoter region is conserved in mosquito species and is responsible for induction of CxVago expression following WNV infection. Using dsRNA-based gene knockdown, we show that the NF-κB ortholog, Rel2, plays significant role in the signaling pathway that activates CxVago in mosquito cells in vitro and in vivo. Using similar approaches, we also show that TRAF, but not TRAF-3, is involved in activation of Rel2 after viral infection. Overall the study shows that a conserved signaling pathway, which is similar to mammalian interferon activation pathway, is responsible for the induction and antiviral activity of CxVago. Viruses like West Nile, dengue and Japanese encephalitis are responsible for large number of human and livestock diseases worldwide. These viruses, transmitted by female mosquitoes via saliva during blood-feeding, elicit an immune response in these mosquitoes. The details of this immune response are still being investigated. Dicer2, which has previously been shown to be involved in RNAi mediated antiviral activity in mosquitoes, contains helicase domain, which leads to activation of antiviral protein, Vago. Vago is functionally similar to mammalian interferon and after secretion activates Jak-STAT pathway in neighboring cells leading to antiviral effect. Here we demonstrate that sensing of viral RNA by Dicer2 leads to activation of TNF receptor-associated factor (TRAF), which in turn leads to cleavage and release of amino-terminal domain of Rel2, NF-κB ortholog. Rel2 binds to a conserved NF-κB binding site on Vago promoter region leading to its induction. This proposed mechanism of Vago activation is similar to mammalian interferon activation after viral infection. The identification of this novel and evolutionarily conserved pathway downstream of Dicer2 provides new insight into the immune signalling in mosquitoes and other invertebrates.
Collapse
Affiliation(s)
- Prasad N. Paradkar
- CSIRO Animal, Food and Health Sciences, Australian Animal Health Laboratory, Geelong, Victoria, Australia
- * E-mail:
| | - Jean-Bernard Duchemin
- CSIRO Ecosystem Sciences, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Rhonda Voysey
- CSIRO Animal, Food and Health Sciences, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Peter J. Walker
- CSIRO Animal, Food and Health Sciences, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| |
Collapse
|
71
|
Stathopoulos S, Neafsey DE, Lawniczak MKN, Muskavitch MAT, Christophides GK. Genetic dissection of Anopheles gambiae gut epithelial responses to Serratia marcescens. PLoS Pathog 2014; 10:e1003897. [PMID: 24603764 PMCID: PMC3946313 DOI: 10.1371/journal.ppat.1003897] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 12/09/2013] [Indexed: 12/29/2022] Open
Abstract
Genetic variation in the mosquito Anopheles gambiae profoundly influences its ability to transmit malaria. Mosquito gut bacteria are shown to influence the outcome of infections with Plasmodium parasites and are also thought to exert a strong drive on genetic variation through natural selection; however, a link between antibacterial effects and genetic variation is yet to emerge. Here, we combined SNP genotyping and expression profiling with phenotypic analyses of candidate genes by RNAi-mediated silencing and 454 pyrosequencing to investigate this intricate biological system. We identified 138 An. gambiae genes to be genetically associated with the outcome of Serratia marcescens infection, including the peptidoglycan recognition receptor PGRPLC that triggers activation of the antibacterial IMD/REL2 pathway and the epidermal growth factor receptor EGFR. Silencing of three genes encoding type III fibronectin domain proteins (FN3Ds) increased the Serratia load and altered the gut microbiota composition in favor of Enterobacteriaceae. These data suggest that natural genetic variation in immune-related genes can shape the bacterial population structure of the mosquito gut with high specificity. Importantly, FN3D2 encodes a homolog of the hypervariable pattern recognition receptor Dscam, suggesting that pathogen-specific recognition may involve a broader family of immune factors. Additionally, we showed that silencing the gene encoding the gustatory receptor Gr9 that is also associated with the Serratia infection phenotype drastically increased Serratia levels. The Gr9 antibacterial activity appears to be related to mosquito feeding behavior and to mostly rely on changes of neuropeptide F expression, together suggesting a behavioral immune response following Serratia infection. Our findings reveal that the mosquito response to oral Serratia infection comprises both an epithelial and a behavioral immune component. In malaria vector mosquitoes, the presence of bacteria and malaria parasites is tightly linked. Bacteria that are part of the mosquito gut ecosystem are critical modulators of the immune response elicited during infection with malaria parasites. Furthermore, responses against oral bacterial infections can affect malaria parasites. Here, we combined mosquito gut infections with the enterobacterium Serratia marcescens with genome-wide discovery and phenotypic analysis of genes involved in antibacterial responses to characterize molecular processes that control gut bacterial infections thus possibly affecting the mosquito susceptibility to infection by malaria parasites. Our data reveal complex genetic networks controlling the gut bacterial infection load and ecosystem homeostasis. These networks appear to exhibit much higher specificity toward specific classes of bacteria than previously thought and include behavioral response circuits involved in antibacterial immunity.
Collapse
Affiliation(s)
| | | | | | | | - George K. Christophides
- Department of Life Sciences, Imperial College London, London, United Kingdom
- The Cyprus Institute, Nicosia, Cyprus
- * E-mail:
| |
Collapse
|
72
|
Zhang M, Zhou F, Chu Y, Zhao Z, An C. Identification and expression profile analysis of antimicrobial peptide/protein in Asian corn borer, Ostrinia furnacalis (Guenée). Int J Biol Sci 2013; 9:1004-12. [PMID: 24155672 PMCID: PMC3805904 DOI: 10.7150/ijbs.6813] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/21/2013] [Indexed: 12/20/2022] Open
Abstract
Antimicrobial peptides/proteins (AMPs) are a group of immune proteins that exhibit strong antibiotic properties against numerous infectious bacterial strains. They are evolutionarily conserved and present in every kingdom and phylum, ranging from prokaryotes to humans. We analyzed the transciptome from the larvae of Asian corn borer, Ostrinia furnacalis (Guenée), and identified several putative AMP transcripts, OfgLys5, OfgLys6, OfgLys10, OfgAtt, and OfgIID. OfgLys5, OfgLys6, and OfgLys10 are all highly homologous with c-type lysozymes, and OfgAtt shows significant identities with Lepidoptera attacin. The amino acid sequence of OfgLys5 and OfgLys6 possessed all conserved features critical for fundamental structure and function of c-type lysozyme, including the two catalytic sites, Glu32 and Asp50. OfgAtt is a typical glycine-rich protein. The antimicrobial activity of O. furnacalis hemolymph increased significantly after injection with Escherichia coli, Micrococcus luteus, or Beauveria bassiana. OfgAtt, IDD, and Lys6 are expressed at low level prior to the challenge, but strongly induced against Gram-positive and negative bacteria, and fungi. Under the same inducement conditions, the transcripts of these three genes elevated most when fifth instar larvae were injected. Therefore, O. furnacalis larvae are induced to produce antimicrobial materials in the hemolymph after the infection, and increase of lysozyme and attacin may contribute to the antimicrobial activity.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Entomology, China Agricultural University, Beijing 100193, P. R. China
| | | | | | | | | |
Collapse
|
73
|
The developmental transcriptome of the mosquito Aedes aegypti, an invasive species and major arbovirus vector. G3-GENES GENOMES GENETICS 2013; 3:1493-509. [PMID: 23833213 PMCID: PMC3755910 DOI: 10.1534/g3.113.006742] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mosquitoes are vectors of a number of important human and animal diseases. The development of novel vector control strategies requires a thorough understanding of mosquito biology. To facilitate this, we used RNA-seq to identify novel genes and provide the first high-resolution view of the transcriptome throughout development and in response to blood feeding in a mosquito vector of human disease, Aedes aegypti, the primary vector for Dengue and yellow fever. We characterized mRNA expression at 34 distinct time points throughout Aedes development, including adult somatic and germline tissues, by using polyA+ RNA-seq. We identify a total of 14,238 novel new transcribed regions corresponding to 12,597 new loci, as well as many novel transcript isoforms of previously annotated genes. Altogether these results increase the annotated fraction of the transcribed genome into long polyA+ RNAs by more than twofold. We also identified a number of patterns of shared gene expression, as well as genes and/or exons expressed sex-specifically or sex-differentially. Expression profiles of small RNAs in ovaries, early embryos, testes, and adult male and female somatic tissues also were determined, resulting in the identification of 38 new Aedes-specific miRNAs, and ~291,000 small RNA new transcribed regions, many of which are likely to be endogenous small-interfering RNAs and Piwi-interacting RNAs. Genes of potential interest for transgene-based vector control strategies also are highlighted. Our data have been incorporated into a user-friendly genome browser located at www.Aedes.caltech.edu, with relevant links to Vectorbase (www.vectorbase.org)
Collapse
|
74
|
Sreenivasamurthy SK, Dey G, Ramu M, Kumar M, Gupta MK, Mohanty AK, Harsha HC, Sharma P, Kumar N, Pandey A, Kumar A, Prasad TSK. A compendium of molecules involved in vector-pathogen interactions pertaining to malaria. Malar J 2013; 12:216. [PMID: 23802619 PMCID: PMC3734095 DOI: 10.1186/1475-2875-12-216] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 06/24/2013] [Indexed: 02/06/2023] Open
Abstract
Malaria is a vector-borne disease causing extensive morbidity, debility and mortality. Development of resistance to drugs among parasites and to conventional insecticides among vector-mosquitoes necessitates innovative measures to combat this disease. Identification of molecules involved in the maintenance of complex developmental cycles of the parasites within the vector and the host can provide attractive targets to intervene in the disease transmission. In the last decade, several efforts have been made in identifying such molecules involved in mosquito-parasite interactions and, subsequently, validating their role in the development of parasites within the vector. In this study, a list of mosquito proteins, which facilitate or inhibit the development of malaria parasites in the midgut, haemolymph and salivary glands of mosquitoes, is compiled. A total of 94 molecules have been reported and validated for their role in the development of malaria parasites inside the vector. This compendium of molecules will serve as a centralized resource to biomedical researchers investigating vector-pathogen interactions and malaria transmission.
Collapse
|
75
|
Juvenile hormone and its receptor, methoprene-tolerant, control the dynamics of mosquito gene expression. Proc Natl Acad Sci U S A 2013; 110:E2173-81. [PMID: 23633570 DOI: 10.1073/pnas.1305293110] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Juvenile hormone III (JH) plays a key role in regulating the reproduction of female mosquitoes. Microarray time-course analysis revealed dynamic changes in gene expression during posteclosion (PE) development in the fat body of female Aedes aegypti. Hierarchical clustering identified three major gene clusters: 1,843 early-PE (EPE) genes maximally expressed at 6 h PE, 457 mid-PE (MPE) genes at 24 h PE, and 1,815 late-PE (LPE) genes at 66 h PE. The RNAi microarray screen for the JH receptor Methoprene-tolerant (Met) showed that 27% of EPE and 40% of MPE genes were up-regulated whereas 36% of LPE genes were down-regulated in the absence of this receptor. Met repression of EPE and MPE and activation of LPE genes were validated by an in vitro fat-body culture experiment using Met RNAi. Sequence motif analysis revealed the consensus for a 9-mer Met-binding motif, CACG(C)/TG(A)/G(T)/AG. Met-binding motif variants were overrepresented within the first 300 bases of the promoters of Met RNAi-down-regulated (LPE) genes but not in Met RNAi-up-regulated (EPE) genes. EMSAs using a combination of mutational and anti-Met antibody supershift analyses confirmed the binding properties of the Met consensus motif variants. There was a striking temporal separation of expression profiles among major functional gene groups, with carbohydrate, lipid, and xenobiotics metabolism belonging to the EPE and MPE clusters and transcription and translation to the LPE cluster. This study represents a significant advancement in the understanding of the regulation of gene expression by JH and its receptor Met during female mosquito reproduction.
Collapse
|
76
|
Gulley MM, Zhang X, Michel K. The roles of serpins in mosquito immunology and physiology. JOURNAL OF INSECT PHYSIOLOGY 2013; 59:138-47. [PMID: 22960307 PMCID: PMC3560325 DOI: 10.1016/j.jinsphys.2012.08.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/24/2012] [Accepted: 08/28/2012] [Indexed: 05/10/2023]
Abstract
In vector-borne diseases, the complex interplay between pathogen and its vector's immune system determines the outcome of infection and therefore disease transmission. Serpins have been shown in many animals to be key regulators of innate immune reactions. Their control over regulatory proteolytic cascades ultimately decides whether the recognition of a pathogen will lead to an appropriate immune response. In mosquitoes, serpins (SRPNs) regulate the activation of prophenoloxidase and thus melanization, contribute to malaria parasite lysis, and likely Toll pathway activation. Additionally, in culicine mosquitoes, SRPNs are able to regulate hemostasis in the vertebrate host, suggesting a crucial role during bloodfeeding. This review summarizes the annotation, transcriptional regulation, and current knowledge of SRPN function in the three mosquito species for which the complete genome sequence is available. Additionally, we give a brief overview of how SRPNs may be used to prevent transmission of vector-borne diseases.
Collapse
Affiliation(s)
| | | | - Kristin Michel
- Corresponding author: tel.: +1 (785) 532-0161, fax: +1 (785) 532-6653;
| |
Collapse
|
77
|
Altered immunity in crowded locust reduced fungal (Metarhizium anisopliae) pathogenesis. PLoS Pathog 2013; 9:e1003102. [PMID: 23326229 PMCID: PMC3542111 DOI: 10.1371/journal.ppat.1003102] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 11/09/2012] [Indexed: 01/16/2023] Open
Abstract
The stress of living conditions, similar to infections, alters animal immunity. High population density is empirically considered to induce prophylactic immunity to reduce the infection risk, which was challenged by a model of low connectivity between infectious and susceptible individuals in crowded animals. The migratory locust, which exhibits polyphenism through gregarious and solitary phases in response to population density and displays different resistance to fungal biopesticide (Metarhizium anisopliae), was used to observe the prophylactic immunity of crowded animals. We applied an RNA-sequencing assay to investigate differential expression in fat body samples of gregarious and solitary locusts before and after infection. Solitary locusts devoted at least twice the number of genes for combating M. anisopliae infection than gregarious locusts. The transcription of immune molecules such as pattern recognition proteins, protease inhibitors, and anti-oxidation proteins, was increased in prophylactic immunity of gregarious locusts. The differentially expressed transcripts reducing gregarious locust susceptibility to M. anisopliae were confirmed at the transcriptional and translational level. Further investigation revealed that locust GNBP3 was susceptible to proteolysis while GNBP1, induced by M. anisopliae infection, resisted proteolysis. Silencing of gnbp3 by RNAi significantly shortened the life span of gregarious locusts but not solitary locusts. By contrast, gnbp1 silencing did not affect the life span of both gregarious and solitary locusts after M. anisopliae infection. Thus, the GNBP3-dependent immune responses were involved in the phenotypic resistance of gregarious locusts to fungal infection, but were redundant in solitary locusts. Our results indicated that gregarious locusts prophylactically activated upstream modulators of immune cascades rather than downstream effectors, preferring to quarantine rather than eliminate pathogens to conserve energy meanwhile increasing the "distance" of infectious and target individuals. Our study has obvious implications for bio-pesticides management of crowded pests, and for understanding disease epidemics and adaptiveness of pathogens.
Collapse
|
78
|
Immune transcript variations among Aedes aegypti populations with distinct susceptibility to dengue virus serotype 2. Acta Trop 2012; 124:113-9. [PMID: 22877626 DOI: 10.1016/j.actatropica.2012.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 07/06/2012] [Accepted: 07/09/2012] [Indexed: 01/18/2023]
Abstract
The innate immune response of insects is one of the factors that may dictate their susceptibility to viral infection. Two immune signaling pathways, Toll and JAK-STAT, and the RNA interference (RNAi) pathway are involved in Aedes aegypti responses against dengue virus (DENV), however natural differences in these antiviral defenses among mosquito populations have not been studied. Here, two field Ae. aegypti populations from distinct ecological environments, one from Recife and the other from Petrolina (Brazil), and a laboratory strain were studied for their ability to replicate a primary isolate of dengue virus serotype 2 (DENV-2). Virus infectivity and replication were determined in insect tissues collected after viral exposure through reverse-transcription real time PCR (RT-PCR). The expression of a transcript representing these defense mechanisms (Toll, JAK-STAT and RNAi) in the midgut and fat body was studied with RT-PCR to evaluate variations in innate immune mechanisms possibly employed against DENV. Analyses of infection rates indicated that the field populations were more susceptible to DENV-2 infection than the lab strain. There were distinct expression patterns among mosquito populations, in both control and infected insects. Moreover, lower expression of immune molecules in DENV-2-infected insects compared to controls was observed in the two field populations. These results suggest that natural variations in vector competence against DENV may be partly due to differences in mosquito defense mechanisms, and that the down-regulation of immune transcripts after viral infection depends on the insect strain.
Collapse
|
79
|
Caljon G, De Vooght L, Van Den Abbeele J. Options for the delivery of anti-pathogen molecules in arthropod vectors. J Invertebr Pathol 2012; 112 Suppl:S75-82. [PMID: 22841635 DOI: 10.1016/j.jip.2012.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 05/15/2012] [Accepted: 05/18/2012] [Indexed: 10/28/2022]
Abstract
Blood feeding arthropods are responsible for the transmission of a large array of medically important infectious agents that include viruses, bacteria, protozoan parasites and helminths. The recent development of transgenic and paratransgenic technologies have enabled supplementing the immune system of these arthropod vectors with anti-pathogen effector molecules in view of compromising their vector competence for these microbial agents. The characteristics of the selected anti-pathogen compound will largely determine the efficacy and specificity of this approach. Low specificity will generally result in bystander effects, likely having a direct or indirect fitness cost for the arthropod. In contrast, the use of highly specific compounds from the adaptive immune system of vertebrates such as antibody derived fragments is more likely to enable highly specific effects without conferring a selective disadvantage to the (para)transgenic arthropods. Here, Nanobodies® are excellent candidates to increase the immune competence of arthropods. Moreover they were shown to exert a novel type of anti-pathogen activity that uniquely depends on their small size.
Collapse
Affiliation(s)
- Guy Caljon
- Department of Biomedical Sciences, Unit of Veterinary Protozoology, Institute of Tropical Medicine Antwerp (ITM), Antwerp, Belgium.
| | | | | |
Collapse
|
80
|
Donald CL, Kohl A, Schnettler E. New Insights into Control of Arbovirus Replication and Spread by Insect RNA Interference Pathways. INSECTS 2012; 3:511-31. [PMID: 26466541 PMCID: PMC4553608 DOI: 10.3390/insects3020511] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 05/11/2012] [Accepted: 05/16/2012] [Indexed: 12/17/2022]
Abstract
Arthropod-borne (arbo) viruses are transmitted by vectors, such as mosquitoes, to susceptible vertebrates. Recent research has shown that arbovirus replication and spread in mosquitoes is not passively tolerated but induces host responses to control these pathogens. Small RNA-mediated host responses are key players among these antiviral immune strategies. Studies into one such small RNA-mediated antiviral response, the exogenous RNA interference (RNAi) pathway, have generated a wealth of information on the functions of this mechanism and the enzymes which mediate antiviral activities. However, other small RNA-mediated host responses may also be involved in modulating antiviral activity. The aim of this review is to summarize recent research into the nature of small RNA-mediated antiviral responses in mosquitoes and to discuss future directions for this relatively new area of research.
Collapse
Affiliation(s)
- Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, UK.
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, UK.
| | - Esther Schnettler
- MRC-University of Glasgow Centre for Virus Research, 8 Church Street, Glasgow G11 5JR, Scotland, UK.
| |
Collapse
|