51
|
Limthongkul J, Mapratiep N, Apichirapokey S, Suksatu A, Midoeng P, Ubol S. Insect anionic septapeptides suppress DENV replication by activating antiviral cytokines and miRNAs in primary human monocytes. Antiviral Res 2019; 168:1-8. [PMID: 31075349 DOI: 10.1016/j.antiviral.2019.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/10/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
Dengue viruses (DENVs) have threatened 2/3 of the world population for decades. Thus, combating DENV infection with either antiviral therapy or protective vaccination is an urgent goal. In the present study, we investigated the anti-DENV activity of insect cell-derived anionic septapeptides from C6/36 mosquito cell cultures persistently infected with DENV. These molecules were previously shown to protect C6/36 and Vero cells against DENV infection. We found that treatment with these septapeptides strongly and rapidly downregulated the multiplication of DENV-1 16007, DENV-3 16562, and DENV-4 1036 but not that of DENV-2 16681 in primary human monocytes. This inhibitory effect was likely mediated through various routes including the increased production of antiviral cytokines (IFN-I), activation of mononuclear cell migration, and upregulation of the expression of antiviral miRNAs (has-miR-30e*, has-miR-133a, and has-miR-223) and inflammation-related miRNAs (has-miR-146a and has-miR-147). In conclusion, anionic septapeptides exerted anti-DENV activity in human monocytes through the upregulation of innate immune responses and the activation of several previously reported antiviral and inflammation-related miRNAs.
Collapse
Affiliation(s)
- Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Nithipong Mapratiep
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Suttikarn Apichirapokey
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Ampa Suksatu
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Panuwat Midoeng
- Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand.
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| |
Collapse
|
52
|
Lee WS, Webster JA, Madzokere ET, Stephenson EB, Herrero LJ. Mosquito antiviral defense mechanisms: a delicate balance between innate immunity and persistent viral infection. Parasit Vectors 2019; 12:165. [PMID: 30975197 PMCID: PMC6460799 DOI: 10.1186/s13071-019-3433-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/04/2019] [Indexed: 01/24/2023] Open
Abstract
Mosquito-borne diseases are associated with major global health burdens. Aedes spp. and Culex spp. are primarily responsible for the transmission of the most medically important mosquito-borne viruses, including dengue virus, West Nile virus and Zika virus. Despite the burden of these pathogens on human populations, the interactions between viruses and their mosquito hosts remain enigmatic. Viruses enter the midgut of a mosquito following the mosquito’s ingestion of a viremic blood meal. During infection, virus recognition by the mosquito host triggers their antiviral defense mechanism. Of these host defenses, activation of the RNAi pathway is the main antiviral mechanism, leading to the degradation of viral RNA, thereby inhibiting viral replication and promoting viral clearance. However, whilst antiviral host defense mechanisms limit viral replication, the mosquito immune system is unable to effectively clear the virus. As such, these viruses can establish persistent infection with little or no fitness cost to the mosquito vector, ensuring life-long transmission to humans. Understanding of the mosquito innate immune response enables the discovery of novel antivectorial strategies to block human transmission. This review provides an updated and concise summary of recent studies on mosquito antiviral immune responses, which is a key determinant for successful virus transmission. In addition, we will also discuss the factors that may contribute to persistent infection in mosquito hosts. Finally, we will discuss current mosquito transmission-blocking strategies that utilize genetically modified mosquitoes and Wolbachia-infected mosquitoes for resistance to pathogens.
Collapse
Affiliation(s)
- Wai-Suet Lee
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Julie A Webster
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Eugene T Madzokere
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Eloise B Stephenson
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia.,Environmental Futures Research Institute, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD, 4215, Australia.
| |
Collapse
|
53
|
Homologs of Human Dengue-Resistance Genes, FKBP1B and ATCAY, Confer Antiviral Resistance in Aedes aegypti Mosquitoes. INSECTS 2019; 10:insects10020046. [PMID: 30717390 PMCID: PMC6409984 DOI: 10.3390/insects10020046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023]
Abstract
Dengue virus (DENV) is transmitted by mosquitoes and is a major public health concern. The study of innate mosquito defense mechanisms against DENV have revealed crucial roles for the Toll, Imd, JAK-STAT, and RNAi pathways in mediating DENV in the mosquito. Often overlooked in such studies is the role of intrinsic cellular defense mechanisms that we hypothesize to work in concert with the classical immune pathways to affect organismal defense. Our understanding of the molecular interaction of DENV with mosquito host cells is limited, and we propose to expand upon the recent results from a genome-scale, small interfering RNA (siRNA)-based study that identified mammalian host proteins associated with resistance to dengue/West Nile virus (DENV/WNV) infection. The study identified 22 human DENV/WNV resistance genes (DVR), and we hypothesized that a subset would be functionally conserved in Aedes aegypti mosquitoes, imparting cellular defense against flaviviruses in this species. We identified 12 homologs of 22 human DVR genes in the Ae. aegypti genome. To evaluate their possible role in cellular resistance/antiviral defense against DENV, we used siRNA silencing targeted against each of the 12 homologs in an Ae. aegypti cell line (Aag2) infected with DENV2 and identified that silencing of the two candidates, AeFKBP1 and AeATCAY, homologs of human FKBP1B and ATCAY, were associated with a viral increase. We then used dsRNA to silence each of the two genes in adult mosquitoes to validate the observed antiviral functions in vivo. Depletion of AeFKBP1 or AeATCAY increased viral dissemination through the mosquito at 14 days post-infection. Our results demonstrated that AeFKBP1 and AeATCAY mediate resistance to DENV akin to what has been described for their homologs in humans. AeFKBP1 and AeATCAY provide a rare opportunity to elucidate a DENV-resistance mechanism that may be evolutionarily conserved between humans and Ae. aegypti.
Collapse
|
54
|
Huang YJS, Higgs S, Vanlandingham DL. Arbovirus-Mosquito Vector-Host Interactions and the Impact on Transmission and Disease Pathogenesis of Arboviruses. Front Microbiol 2019; 10:22. [PMID: 30728812 PMCID: PMC6351451 DOI: 10.3389/fmicb.2019.00022] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022] Open
Abstract
Hundreds of viruses, designated as arboviruses, are transmitted by arthropod vectors in complex transmission cycles between the virus, vertebrate host, and the vector. With millions of human and animal infections per year, it is critical to improve our understanding of the interactions between the biological and environmental factors that play a critical role in pathogenesis, disease outcomes, and transmission of arboviruses. This review focuses on mosquito-borne arboviruses and discusses current knowledge of the factors and underlying mechanisms that influence infection and transmission of arboviruses and discusses critical factors and pathways that can potentially become targets for intervention and therapeutics.
Collapse
Affiliation(s)
- Yan-Jang S Huang
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| | - Stephen Higgs
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| | - Dana L Vanlandingham
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
55
|
Wei L, Yang Y, Zhou Y, Li M, Yang H, Mu L, Qian Q, Wu J, Xu W. Anti-inflammatory activities of Aedes aegypti cecropins and their protection against murine endotoxin shock. Parasit Vectors 2018; 11:470. [PMID: 30107813 PMCID: PMC6092832 DOI: 10.1186/s13071-018-3000-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 07/06/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mosquitoes are armed with physiologically active compounds to suppress the host immunity including host inflammatory reaction. However, the specific anti-inflammatory components in mosquitoes remain unknown. RESULTS By searching for the immunomodulatory molecules from the mosquito Aedes aegypti (Diptera: Culicidae) at NCBI for anti-inflammatory function, five cecropins (for short in this study: AeaeCec1, 2, 3, 4 and 5) were selected. AeaeCec1-5 efficiently inhibited the expression of inducible nitric oxide synthase (iNOS), nitrite, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and interleukin-6 (IL-6) in lipopolysaccharide (LPS)-stimulated mouse peritoneal macrophages and human peripheral blood mononuclear cells (PBMCs) with low toxicity to mammalian cells. Among the five analogues, AeaeCec5 had the strongest anti-inflammatory activity, and generated an additive effect with other AeaeCec peptides. In a mouse model of endotoxin shock, AeaeCec1-5 effectively reduced TNF-α, IL-1β and IL-6 expression in lungs, serum and peritoneal lavage and correspondingly reduced lung damage and edema, with AeaeCec5 showing the best protection. In mice infected with Escherichia coli or Pseudomonas aeruginosa, administration of AeaeCec5 reduced the production of TNF-α, IL-1β and IL-6 and correspondingly reduced lung tissue damage. These effects of Ae. aegypti AeaeCec1-5 were attributed to an efficient inhibition of the activation of mitogen-activated protein kinases (MAPKs) and transcriptional nuclear factor-κB (NF-κB) signaling pathways, as well as partial neutralization of LPS. CONCLUSIONS The current work characterized the specific anti-inflammatory agents in Ae. aegypti and provided AeaeCec5 as a potent anti-endotoxin peptide that could serve as the basis for the development of anti-inflammatory therapy.
Collapse
Affiliation(s)
- Lin Wei
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yang Yang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yandong Zhou
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Min Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hailong Yang
- School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Lixian Mu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, Yunnan, China
| | - Qian Qian
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Jing Wu
- School of Basic Medical Sciences, Kunming Medical University, Kunming, 650500, Yunnan, China.
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
56
|
Shrinet J, Bhavesh NS, Sunil S. Understanding Oxidative Stress in Aedes during Chikungunya and Dengue Virus Infections Using Integromics Analysis. Viruses 2018; 10:v10060314. [PMID: 29890729 PMCID: PMC6024870 DOI: 10.3390/v10060314] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
Arboviral infection causes dysregulation of cascade of events involving numerous biomolecules affecting fitness of mosquito to combat virus. In response of the viral infection mosquito’s defense mechanism get initiated. Oxidative stress is among the first host responses triggered by the vector. Significant number of information is available showing changes in the transcripts and/or proteins upon Chikungunya virus and Dengue virus mono-infections and as co-infections. In the present study, we collected different -omics data available in the public database along with the data generated in our laboratory related to mono-infections or co-infections of these viruses. We analyzed the data and classified them into their respective pathways to study the role of oxidative stress in combating arboviral infection in Aedes mosquito. The analysis revealed that the oxidative stress related pathways functions in harmonized manner.
Collapse
Affiliation(s)
- Jatin Shrinet
- Vector Borne Diseases, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Neel Sarovar Bhavesh
- Transcriptional Regulation, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Sujatha Sunil
- Vector Borne Diseases, International Centre for Genetic Engineering and Biotechnology (ICGEB), Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
57
|
Wang J, Song X, Wang M. Peptidoglycan recognition proteins in hematophagous arthropods. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:89-95. [PMID: 29269264 PMCID: PMC5889321 DOI: 10.1016/j.dci.2017.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 05/24/2023]
Abstract
Hematophagous arthropods are medically important disease vectors that transmit a variety of pathogens. Unlike mammals that employ both innate and adaptive immunity to clear invading pathogens, these vectors rely mainly on an innate immune system to combat pathogens. Peptidoglycan recognition proteins (PGRPs) are important components of innate immune signaling pathways and are responsible for recognizing microbe-associated molecular patterns (MAMPs), thus regulating host immune interactions with both harmful and helpful microbes. Here we review a number of recent studies in different vectors that address the function of PGRPs in immune regulation. Further, we discuss the variation of PGRPs between vectors and Drosophila.
Collapse
Affiliation(s)
- Jingwen Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China.
| | - Xiumei Song
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| | - Mengfei Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| |
Collapse
|
58
|
Liu T, Xu Y, Wang X, Gu J, Yan G, Chen XG. Antiviral systems in vector mosquitoes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:34-43. [PMID: 29294302 DOI: 10.1016/j.dci.2017.12.025] [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: 09/27/2017] [Revised: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 06/07/2023]
Abstract
Mosquito-borne viral diseases represent a major challenge to human public health. As natural vectors of arboviruses, mosquitoes can be infected by a virus, but they have evolved multiple mechanisms to tolerate constant infection and restrict viral replication via their antiviral immune system. In a state of continuous infection, a mosquito can transmit an arbovirus while obtaining a blood meal from a mammalian host. During infection, the virus is mainly inhibited through a small RNA-mediated interference mechanism. Within mosquitoes, the invaded viruses are recognized based on pathogen-associated molecular patterns, leading to the production of cytokines. These cytokines in turn bind pattern recognition receptors and activate Toll, IMD and other immune signalling pathways to expand the immune response and induce antiviral activity via immune effectors. Interestingly, the gut microbiota and Wolbachia also play a role in mosquito antiviral immunity, which is very similar to acquired immunity. This review describes the advances made in understanding various aspects of mosquito antiviral immune molecular mechanisms in detail and explores some of the unresolved issues related to the mosquito immune system.
Collapse
Affiliation(s)
- Tong Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ye Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoming Wang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinbao Gu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guiyun Yan
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China; Program in Public Health, School of Medicine, University of California, Irvine, USA
| | - Xiao-Guang Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
59
|
Tham HW, Balasubramaniam V, Ooi MK, Chew MF. Viral Determinants and Vector Competence of Zika Virus Transmission. Front Microbiol 2018; 9:1040. [PMID: 29875751 PMCID: PMC5974093 DOI: 10.3389/fmicb.2018.01040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/02/2018] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV) has emerged as a new global health threat. Since its first discovery in Zika forest in Uganda, this virus has been isolated from several mosquito species, including Aedes aegypti and Aedes albopictus. The geographical distribution of these mosquito species across tropical and subtropical regions has led to several outbreaks, including the recent pandemic in Brazil, followed by the Pacific islands and other areas of North and South America. This has gained attention of the scientific community to elucidate the epidemiology and transmission of ZIKV. Despite its strong attention on clinical aspects for healthcare professionals, the relationships between ZIKV and its principal vectors, A. aegypti and A. albopictus, have not gained substantial interest in the scientific research community. As such, this review aims to summarize the current knowledge on ZIKV tropism and some important mechanisms which may be employed by the virus for effective strategies on viral survival in mosquitoes. In addition, this review identifies the areas of research that should be placed attention to, for which to be exploited for novel mosquito control strategies.
Collapse
Affiliation(s)
- Hong-Wai Tham
- Biology Research Laboratory, Faculty of Pharmacy, SEGi University, Petaling Jaya, Malaysia
| | - Vinod Balasubramaniam
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Man K. Ooi
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Miaw-Fang Chew
- Centre for Virus and Vaccine Research, School of Science and Technology, Sunway University, Subang Jaya, Malaysia
| |
Collapse
|
60
|
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
|
61
|
Encapsidated Host Factors in Alphavirus Particles Influence Midgut Infection of Aedes aegypti. Viruses 2018; 10:v10050263. [PMID: 29772674 PMCID: PMC5977256 DOI: 10.3390/v10050263] [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: 04/10/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 12/25/2022] Open
Abstract
Transmission of mosquito-borne viruses requires the efficient infection of both a permissive vertebrate host and a competent mosquito vector. The infectivity of Sindbis virus (SINV), the type species of the Alphavirus genus, is influenced by both the original and new host cell. We have shown that infection of vertebrate cells by SINV, chikungunya virus (CHIKV), and Ross River virus (RRV) produces two subpopulations of virus particles separable based on density. In contrast, a single population of viral particles is produced by mosquito cells. Previous studies demonstrated that the denser vertebrate-derived particles and the mosquito-derived particles contain components of the small subunit of the host cell ribosome, whereas the less dense vertebrate-derived particles do not. Infection of mice with RRV showed that both particle subpopulations are produced in an infected vertebrate, but in a tissue specific manner with serum containing only the less dense version of the virus particles. Previous infectivity studies using SINV particles have shown that the denser particles (SINVHeavy) and mosquito derived particles SINVC6/36 are significantly more infectious in vertebrate cells than the less dense vertebrate derived particles (SINVLight). The current study shows that SINVLight particles, initiate the infection of the mosquito midgut more efficiently than SINVHeavy particles and that this enhanced infectivity is associated with an exacerbated immune response to SINVLight infection in midgut tissues. The enhanced infection of SINVLight is specific to the midgut as intrathoracically injected virus do not exhibit the same fitness advantage. Together, our data indicate a biologically significant role for the SINVLight subpopulation in the efficient transmission from infected vertebrates to the mosquito vector.
Collapse
|
62
|
Gomes KAGG, Dos Santos DM, Santos VM, Piló-Veloso D, Mundim HM, Rodrigues LV, Lião LM, Verly RM, de Lima ME, Resende JM. NMR structures in different membrane environments of three ocellatin peptides isolated from Leptodactylus labyrinthicus. Peptides 2018; 103:72-83. [PMID: 29596881 DOI: 10.1016/j.peptides.2018.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 10/17/2022]
Abstract
The peptides ocellatin-LB1, -LB2 and -F1 have previously been isolated from anurans of the Leptodactylus genus and the sequences are identical from residue 1-22, which correspond to ocellatin-LB1 sequence (GVVDILKGAAKDIAGHLASKVM-NH2), whereas ocellatin-LB2 carries an extra N and ocellatin-F1 extra NKL residues at their C-termini. These peptides showed different spectra of activities and biophysical investigations indicated a direct correlation between membrane-disruptive properties and antimicrobial activities, i.e. ocellatin-F1 > ocellatin-LB1 > ocellatin-LB2. To better characterize their membrane interactions, we report here the detailed three-dimensional NMR structures of these peptides in TFE-d2:H2O (60:40) and in the presence of zwitterionic DPC-d38 and anionic SDS-d25 micellar solutions. Although the three peptides showed significant helical contents in the three mimetic environments, structural differences were noticed. When the structures of the three peptides in the presence of DPC-d38 micelles are compared to each other, a more pronounced curvature is observed for ocellatin-F1 and the bent helix, with the concave face composed mostly of hydrophobic residues, is consistent with the micellar curvature and the amphipathic nature of the molecule. Interestingly, an almost linear helical segment was observed for ocellatin-F1 in the presence of SDS-d25 micelles and the conformational differences in the two micellar environments are possibly related to the presence of the extra Lys residue near the peptide C-terminus, which increases the affinity of ocellatin-F1 to anionic membranes in comparison with ocellatin-LB1 and -LB2, as proved by isothermal titration calorimetry. To our knowledge, this work reports for the first time the three-dimensional structures of ocellatin peptides.
Collapse
Affiliation(s)
- Karla A G G Gomes
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil; Instituto de Engenharia, Ciência e Tecnologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, 39440-000 Janaúba, MG, Brazil
| | - Daniel M Dos Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Virgílio M Santos
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Dorila Piló-Veloso
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Higor M Mundim
- Instituto de Química, Universidade Federal de Goiás, Av. Esperança, s/n, Campus Samambaia, 74690-900 Goiânia, GO, Brazil
| | - Leticia V Rodrigues
- Instituto de Química, Universidade Federal de Goiás, Av. Esperança, s/n, Campus Samambaia, 74690-900 Goiânia, GO, Brazil
| | - Luciano M Lião
- Instituto de Química, Universidade Federal de Goiás, Av. Esperança, s/n, Campus Samambaia, 74690-900 Goiânia, GO, Brazil
| | - Rodrigo M Verly
- Departamento de Química, Universidade Federal dos Vales do Jequitinhonha e Mucuri, 39100-000 Diamantina, MG, Brazil
| | - Maria Elena de Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil
| | - Jarbas M Resende
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, P.O. Box 486, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
63
|
Samuel GH, Adelman ZN, Myles KM. Antiviral Immunity and Virus-Mediated Antagonism in Disease Vector Mosquitoes. Trends Microbiol 2018; 26:447-461. [PMID: 29395729 PMCID: PMC5910197 DOI: 10.1016/j.tim.2017.12.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022]
Abstract
More than 100 pathogens, spanning multiple virus families, broadly termed 'arthropod-borne viruses (arboviruses)' have been associated with human and/or animal diseases. These viruses persist in nature through transmission cycles that involve alternating replication in susceptible vertebrate and invertebrate hosts. Collectively, these viruses are among the greatest burdens to global health, due to their widespread prevalence, and the severe morbidity and mortality they cause in human and animal hosts. Specific examples of mosquito-borne pathogens include Zika virus (ZIKV), West Nile virus (WNV), dengue virus serotypes 1-4 (DENV 1-4), Japanese encephalitis virus (JEV), yellow fever virus (YFV), chikungunya virus (CHIKV), and Rift Valley fever virus (RVFV). Interactions between arboviruses and the immune pathways of vertebrate hosts have been extensively reviewed. In this review we focus on the antiviral immune pathways present in mosquitoes. We also discuss mechanisms by which mosquito-borne viruses may antagonize antiviral pathways in disease vectors. Finally, we elaborate on the possibility that mosquito-borne viruses may be engaged in an evolutionary arms race with their invertebrate vector hosts, and the possible implications of this for understanding the transmission of mosquito-borne viruses.
Collapse
Affiliation(s)
- Glady Hazitha Samuel
- Texas A & M University, Department of Entomology, Minnie Belle Heep Center, College Station, TX 77843-2475, USA
| | - Zach N Adelman
- Texas A & M University, Department of Entomology, Minnie Belle Heep Center, College Station, TX 77843-2475, USA
| | - Kevin M Myles
- Texas A & M University, Department of Entomology, Minnie Belle Heep Center, College Station, TX 77843-2475, USA.
| |
Collapse
|
64
|
Subverting the mechanisms of cell death: flavivirus manipulation of host cell responses to infection. Biochem Soc Trans 2018; 46:609-617. [DOI: 10.1042/bst20170399] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/15/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
Viruses exploit host metabolic and defence machinery for their own replication. The flaviviruses, which include Dengue (DENV), Yellow Fever (YFV), Japanese Encephalitis (JEV), West Nile (WNV) and Zika (ZIKV) viruses, infect a broad range of hosts, cells and tissues. Flaviviruses are largely transmitted by mosquito bites and humans are usually incidental, dead-end hosts, with the notable exceptions of YFV, DENV and ZIKV. Infection by flaviviruses elicits cellular responses including cell death via necrosis, pyroptosis (involving inflammation) or apoptosis (which avoids inflammation). Flaviviruses exploit these mechanisms and subvert them to prolong viral replication. The different effects induced by DENV, WNV, JEV and ZIKV are reviewed. Host cell surface proteoglycans (PGs) bearing glycosaminoglycan (GAG) polysaccharides — heparan/chondroitin sulfate (HS/CS) — are involved in initial flavivirus attachment and during the expression of non-structural viral proteins play a role in disease aetiology. Recent work has shown that ZIKV-infected cells are protected from cell death by exogenous heparin (a GAG structurally similar to host cell surface HS), raising the possibility of further subtle involvement of HS PGs in flavivirus disease processes. The aim of this review is to synthesize information regarding DENV, WNV, JEV and ZIKV from two areas that are usually treated separately: the response of host cells to infection by flaviviruses and the involvement of cell surface GAGs in response to those infections.
Collapse
|
65
|
Abstract
The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology and Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh EH9 3FL UK.
| | - Finny S Varghese
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
66
|
Fukutani KF, Kasprzykowski JI, Paschoal AR, Gomes MDS, Barral A, de Oliveira CI, Ramos PIP, de Queiroz ATL. Meta-Analysis of Aedes aegypti Expression Datasets: Comparing Virus Infection and Blood-Fed Transcriptomes to Identify Markers of Virus Presence. Front Bioeng Biotechnol 2018; 5:84. [PMID: 29376049 PMCID: PMC5768613 DOI: 10.3389/fbioe.2017.00084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/15/2017] [Indexed: 02/05/2023] Open
Abstract
The mosquito Aedes aegypti (L.) is vector of several arboviruses including dengue, yellow fever, chikungunya, and more recently zika. Previous transcriptomic studies have been performed to elucidate altered pathways in response to viral infection. However, the intrinsic coupling between alimentation and infection were unappreciated in these studies. Feeding is required for the initial mosquito contact with the virus and these events are highly dependent. Addressing this relationship, we reinterrogated datasets of virus-infected mosquitoes with two different diet schemes (fed and unfed mosquitoes), evaluating the metabolic cross-talk during both processes. We constructed coexpression networks with the differentially expressed genes of these comparison: virus-infected versus blood-fed mosquitoes and virus-infected versus unfed mosquitoes. Our analysis identified one module with 110 genes that correlated with infection status (representing ~0.7% of the A. aegypti genome). Furthermore, we performed a machine-learning approach and summarized the infection status using only four genes (AAEL012128, AAEL014210, AAEL002477, and AAEL005350). While three of the four genes were annotated as hypothetical proteins, AAEL012128 gene is a membrane amino acid transporter correlated with viral envelope binding. This gene alone is able to discriminate all infected samples and thus should have a key role to discriminate viral infection in the A. aegypti mosquito. Moreover, validation using external datasets found this gene as differentially expressed in four transcriptomic experiments. Therefore, these genes may serve as a proxy of viral infection in the mosquito and the others 106 identified genes provides a framework to future studies.
Collapse
Affiliation(s)
| | - José Irahe Kasprzykowski
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Post-Graduation Program in Biotechnology in Health and Investigative Medicine, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Alexandre Rossi Paschoal
- Federal University of Technology-Paraná, UTFPR, Campus Cornélio Procópio, Cornélio Procópio, Brazil
| | | | - Aldina Barral
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Post-Graduation Program in Health Sciences, School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Camila I de Oliveira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Post-Graduation Program in Health Sciences, School of Medicine, Federal University of Bahia, Salvador, Brazil
| | | | - Artur Trancoso Lopo de Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Post-Graduation Program in Biotechnology in Health and Investigative Medicine, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Post-Graduation Program in Applied Computation, Universida de Estadual de Feira de Santana, Feira de Santana, Brazil
| |
Collapse
|
67
|
Mosquitoes as Arbovirus Vectors: From Species Identification to Vector Competence. PARASITOLOGY RESEARCH MONOGRAPHS 2018. [PMCID: PMC7122353 DOI: 10.1007/978-3-319-94075-5_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mosquitoes and other arthropods transmit a large number of medically important pathogens, in particular viruses. These arthropod-borne viruses (arboviruses) include a wide variety of RNA viruses belonging to the Flaviviridae family (West Nile virus (WNV), Usutu virus (USUV), Dengue virus (DENV), Japanese encephalitis virus (JEV), Zika virus (ZIKV)), the Togaviridae family (Chikungunya virus (CHIKV)), and Bunyavirales order (Rift Valley fever virus (RVFV)) (please refer also to Table 9.1). Arboviral transmission to humans and livestock constitutes a major threat to public health and economy as illustrated by the emergence of ZIKV in the Americas, RVFV outbreaks in Africa, and the worldwide outbreaks of DENV. To answer the question if those viral pathogens also pose a risk to Europe, we need to first answer the key questions (summarized in Fig. 9.1):Who could contribute to such an outbreak? Information about mosquito species resident or imported, potential hosts and viruses able to infect vectors and hosts in Germany is needed. Where would competent mosquito species meet favorable conditions for transmission? Information on the minimum requirements for efficient replication of the virus in a given vector species and subsequent transmission is needed. How do viruses and vectors interact to facilitate transmission? Information on the vector immunity, vector physiology, vector genetics, and vector microbiomes is needed.
Collapse
|
68
|
Liu WT, Tu WC, Lin CH, Yang UC, Chen CC. Involvement of cecropin B in the formation of the Aedes aegypti mosquito cuticle. Sci Rep 2017; 7:16395. [PMID: 29180688 PMCID: PMC5703890 DOI: 10.1038/s41598-017-16625-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/15/2017] [Indexed: 01/13/2023] Open
Abstract
In this study, we found a mosquito antimicrobial peptide (AMP), Aedes aegypti cecropin B (Aacec B), was expressed constitutively in pupae. Knockdown in the pupae of Aacec B using double-stranded RNA (dsRNA) resulted in high mortality, the emergence of deformed adults and an impairment of pharate adult cuticle formation with fewer lamellae being deposited and the helicoidal pattern of the chitin microfibrils being disorganized. Simultaneous injection of Aacec B dsRNA and Aacec B peptide into pupae significantly reduced this mortality and no deformed adults then emerged. The expression levels of Ae. aegypti prophenoloxidase (AaPPO) 3 and AaPPO 4 were significantly reduced in the Aacec B knockdown pupae. Exogenous Aacec B peptide significantly enhanced the transcription of AaPPO 3 in pupae. Knockdown of AaPPO 3 in pupae caused effects similar to Aacec B-knockdown. The Aacec B peptide could be detected in both the cytoplasm and nuclei of pupal cells and was able to bind to the TTGG(A/C)A motif in AaPPO 3 DNA both in vitro and in vivo. These findings suggest that Aacec B plays a crucial role in pharate adult cuticle formation via the regulation of AaPPO 3 gene expression in pupae.
Collapse
Affiliation(s)
- Wei-Ting Liu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Wu-Chun Tu
- Department of Entomology, National Chung Hsing University, Taichung, 402, Taiwan, ROC
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Ueng-Cheng Yang
- Institute of Biomedical Informatics, National Yang-Ming University, National Yang-Ming University, Taipei, 112, Taiwan, ROC
| | - Cheng-Chen Chen
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, 112, Taiwan, ROC.
| |
Collapse
|
69
|
Vogels CB, Göertz GP, Pijlman GP, Koenraadt CJ. Vector competence of European mosquitoes for West Nile virus. Emerg Microbes Infect 2017; 6:e96. [PMID: 29116220 PMCID: PMC5717085 DOI: 10.1038/emi.2017.82] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/23/2017] [Accepted: 08/27/2017] [Indexed: 01/02/2023]
Abstract
West Nile virus (WNV) is an arthropod-borne flavivirus of high medical and veterinary importance. The main vectors for WNV are mosquito species of the Culex genus that transmit WNV among birds, and occasionally to humans and horses, which are ‘dead-end’ hosts. Recently, several studies have been published that aimed to identify the mosquito species that serve as vectors for WNV in Europe. These studies provide insight in factors that can influence vector competence of European mosquito species for WNV. Here, we review the current knowledge on vector competence of European mosquitoes for WNV, and the molecular knowledge on physical barriers, anti-viral pathways and microbes that influence vector competence based on studies with other flaviviruses. By comparing the 12 available WNV vector competence studies with European mosquitoes we evaluate the effect of factors such as temperature, mosquito origin and mosquito biotype on vector competence. In addition, we propose a standardised methodology to allow for comparative studies across Europe. Finally, we identify knowledge gaps regarding vector competence that, once addressed, will provide important insights into WNV transmission and ultimately contribute to effective strategies to control WNV.
Collapse
Affiliation(s)
- Chantal Bf Vogels
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Giel P Göertz
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Constantianus Jm Koenraadt
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| |
Collapse
|
70
|
Zhu Y, Zhang R, Zhang B, Zhao T, Wang P, Liang G, Cheng G. Blood meal acquisition enhances arbovirus replication in mosquitoes through activation of the GABAergic system. Nat Commun 2017; 8:1262. [PMID: 29093445 PMCID: PMC5665997 DOI: 10.1038/s41467-017-01244-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/30/2017] [Indexed: 11/13/2022] Open
Abstract
Mosquitoes are hematophagous insects that carry-on and transmit many human viruses. However, little information is available regarding the common mechanisms underlying the infection of mosquitoes by these viruses. In this study, we reveal that the hematophagous nature of mosquitoes contributes to arboviral infection after a blood meal, which suppresses antiviral innate immunity by activating the GABAergic pathway. dsRNA-mediated interruption of the GABA signaling and blockage of the GABAA receptor by the specific inhibitors both significantly impaired arbovirus replication. Consistently, inoculation of GABA enhanced arboviral infection, indicating that GABA signaling facilitates the arboviral infection of mosquitoes. The ingestion of blood by mosquitoes resulted in robust GABA production from glutamic acid derived from blood protein digestion. The oral introduction of glutamic acid increased virus acquisition by mosquitoes via activation of the GABAergic system. Our study reveals that blood meals enhance arbovirus replication in mosquitoes through activation of the GABAergic system. Transmission of many human viruses depends on replication in their mosquito vectors. Here, Zhu et al. show that glutamic acid digested from the blood meal activates GABA signaling, resulting in suppression of antiviral innate immunity and increased virus replication in mosquitoes.
Collapse
Affiliation(s)
- Yibin Zhu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.,Institute of pathogenic organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China.,School of Life Science, Tsinghua University, Beijing, 100084, China
| | - Rudian Zhang
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.,School of Life Science, Tsinghua University, Beijing, 100084, China
| | - Bei Zhang
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Tongyan Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Penghua Wang
- Department of Microbiology and Immunology, School of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Guodong Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Viral Disease Control and Prevention, Beijing, 102206, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, 310000, China
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China. .,Institute of pathogenic organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
71
|
Zhang G, Asad S, Khromykh AA, Asgari S. Cell fusing agent virus and dengue virus mutually interact in Aedes aegypti cell lines. Sci Rep 2017; 7:6935. [PMID: 28761113 PMCID: PMC5537255 DOI: 10.1038/s41598-017-07279-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 06/26/2017] [Indexed: 12/31/2022] Open
Abstract
The genus Flavivirus contains more than 70 single-stranded, positive-sense arthropod-borne RNA viruses. Some flaviviruses are particularly medically important to humans and other vertebrates including dengue virus (DENV), West Nile virus, and yellow fever virus. These viruses are transmitted to vertebrates by mosquitoes and other arthropod species. Mosquitoes are also infected by insect-specific flaviviruses (ISFs) that do not appear to be infective to vertebrates. Cell fusing agent virus (CFAV) was the first described ISF, which was discovered in an Aedes aegypti cell culture. We found that while CFAV infection could be significantly reduced by application of RNAi against the NS5 gene, removal of the treatment led to quick restoration of CFAV replication. Interestingly, we found that CFAV infection significantly enhanced replication of DENV, and vice versa, DENV infection significantly enhanced replication of CFAV in mosquito cells. We have shown that CFAV infection leads to increase in the expression of ribonuclease kappa (RNASEK), which is known to promote infection of viruses that rely on endocytosis and pH-dependent entry. Knockdown of RNASEK by dsRNA resulted in reduced DENV replication. Thus, increased expression of RNASEK induced by CFAV is likely to contribute to enhanced DENV replication in CFAV-infected cells.
Collapse
Affiliation(s)
- Guangmei Zhang
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sultan Asad
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alexander A Khromykh
- Australian Infectious Disease Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sassan Asgari
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
72
|
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
|
73
|
Wang ZZ, Bing XL, Liu SS, Chen XX. RNA interference of an antimicrobial peptide, Btdef, reduces Tomato yellow leaf curl China virus accumulation in the whitefly Bemisia tabaci. PEST MANAGEMENT SCIENCE 2017; 73:1421-1427. [PMID: 27804213 DOI: 10.1002/ps.4472] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/03/2016] [Accepted: 11/02/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND The whitefly Bemisia tabaci (Gennadius) is considered one of the main pests for agriculture. One important problem with the whitefly is its notorious status as a vector for plant viruses, primarily begomoviruses. We have previously identified a defensin-like antimicrobial peptide, Btdef, from the whitefly B. tabaci MEAM1. However, the function of Btdef in the immune system of the insect vector and begomovirus transmission has yet to be explored. RESULTS To explore the role of Btdef during begomovirus transmission, we firstly investigated the transcriptional response of Btdef following acquisition of Tomato yellow leaf curl China virus (TYLCCNV). The expression of Btdef was up-regulated in the viruliferous whiteflies. After RNA silencing of the Btdef gene in adult whiteflies fed with dsRNA, they were allowed to feed on TYLCCNV-infected plants and then quantified for TYLCCNV DNA titre. Unexpectedly, silencing the Btdef gene reduced both the abundance and expressions of TYLCCNV genes in the whiteflies. In the meantime, the density of the endosymbiont Rickettsia was significantly reduced in dsBtdef-fed whiteflies. CONCLUSION Our data provide evidence that Btdef is involved in begomovirus infection, possibly through symbiont-mediated alteration of begomovirus-whitefly interactions. These findings indicate that Btdef may be targeted for the development of new technology for the control of whitefly-transmitted begomoviruses. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhi-Zhi Wang
- Ministry of Agriculture Key Lab of Agricultural Entomology, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Xiao-Li Bing
- Ministry of Agriculture Key Lab of Agricultural Entomology, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Shu-Sheng Liu
- Ministry of Agriculture Key Lab of Agricultural Entomology, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Xue-Xin Chen
- Ministry of Agriculture Key Lab of Agricultural Entomology, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
74
|
Imipramine Inhibits Chikungunya Virus Replication in Human Skin Fibroblasts through Interference with Intracellular Cholesterol Trafficking. Sci Rep 2017; 7:3145. [PMID: 28600536 PMCID: PMC5466638 DOI: 10.1038/s41598-017-03316-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is an emerging arbovirus of the Togaviridae family that poses a present worldwide threat to human in the absence of any licensed vaccine or antiviral treatment to control viral infection. Here, we show that compounds interfering with intracellular cholesterol transport have the capacity to inhibit CHIKV replication in human skin fibroblasts, a major viral entry site in the human host. Pretreatment of these cells with the class II cationic amphiphilic compound U18666A, or treatment with the FDA-approved antidepressant drug imipramine resulted in a near total inhibition of viral replication and production at the highest concentration used without any cytotoxic effects. Imipramine was found to affect both the fusion and replication steps of the viral life cycle. The key contribution of cholesterol availability to the CHIKV life cycle was validated further by the use of fibroblasts from Niemann-Pick type C (NPC) patients in which the virus was unable to replicate. Interestingly, imipramine also strongly inhibited the replication of several Flaviviridae family members, including Zika, West Nile and Dengue virus. Together, these data show that this compound is a potential drug candidate for anti-arboviral treatment.
Collapse
|
75
|
Mussabekova A, Daeffler L, Imler JL. Innate and intrinsic antiviral immunity in Drosophila. Cell Mol Life Sci 2017; 74:2039-2054. [PMID: 28102430 PMCID: PMC5419870 DOI: 10.1007/s00018-017-2453-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/11/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
The fruit fly Drosophila melanogaster has been a valuable model to investigate the genetic mechanisms of innate immunity. Initially focused on the resistance to bacteria and fungi, these studies have been extended to include antiviral immunity over the last decade. Like all living organisms, insects are continually exposed to viruses and have developed efficient defense mechanisms. We review here our current understanding on antiviral host defense in fruit flies. A major antiviral defense in Drosophila is RNA interference, in particular the small interfering (si) RNA pathway. In addition, complex inducible responses and restriction factors contribute to the control of infections. Some of the genes involved in these pathways have been conserved through evolution, highlighting loci that may account for susceptibility to viral infections in humans. Other genes are not conserved and represent species-specific innovations.
Collapse
Affiliation(s)
- Assel Mussabekova
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France.
| | - Laurent Daeffler
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France
| | - Jean-Luc Imler
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France
- Faculté des Sciences de la Vie, Université de Strasbourg, 28 rue Goethe, 67000, Strasbourg, France
| |
Collapse
|
76
|
Dengue virus replicates and accumulates in Aedes aegypti salivary glands. Virology 2017; 507:75-81. [PMID: 28431281 DOI: 10.1016/j.virol.2017.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/20/2022]
Abstract
Dengue virus (DENV) is an RNA virus transmitted among humans by mosquito vectors, mainly Aedes aegypti. DENV transmission requires viral dissemination from the mosquito midgut to the salivary glands. During this process the virus undergoes several population bottlenecks, which are stochastic reductions in population size that restrict intra-host viral genetic diversity and limit the efficiency of natural selection. Despite the implications for virus transmission and evolution, DENV replication in salivary glands has not been directly demonstrated. Here, we used a strand-specific quantitative RT-PCR assay to demonstrate that negative-strand DENV RNA is produced in Ae. aegypti salivary glands, providing conclusive evidence that viral replication occurs in this tissue. Furthermore, we showed that the concentration of DENV genomic RNA in salivary glands increases significantly over time, indicating that active replication likely replenishes DENV genetic diversity prior to transmission. These findings improve our understanding of the biological determinants of DENV fitness and evolution.
Collapse
|
77
|
Avila-Bonilla RG, Yocupicio-Monroy M, Marchat LA, De Nova-Ocampo MA, Del Ángel RM, Salas-Benito JS. Analysis of the miRNA profile in C6/36 cells persistently infected with dengue virus type 2. Virus Res 2017; 232:139-151. [PMID: 28267608 DOI: 10.1016/j.virusres.2017.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/28/2022]
Abstract
Dengue virus (DENV) is the most important arbovirus in the world; DENV is transmitted by the Aedes genus of mosquitoes and can establish a life-long persistent infection in mosquitoes. However, the exact mechanism by which persistent infection is established remains unknown. In this study the differential expression of miRNAs was analysed by deep sequencing and RT-qPCR using a previously established C6/36-HT cell line persistently infected with DENV 2 (C6-L) as a model. miR-927, miR-87, miR-210, miR-2a-3p, miR-190 and miR-970 were up-regulated, whereas miR-252, miR-263a-3p, miR-92b, miR-10-5p miR-9a-5p, miR-9a-1, miR-124, miR-286a and miR-286b were down-regulated in C6-L cells compared with C6/36 cells acutely infected with the same virus or mock-infected cells. Deep sequencing results were validated by RT-qPCR for the highly differentially expressed miR-927 and miR-9a-5p, which were up- and down-regulated, respectively, compared with both acutely and mock-infected C6/36 cells. The putative targets of these miRNAs include components of the ubiquitin conjugation pathway, vesicle-mediated transport, autophagy, and the JAK-STAT cascade as well as proteins with endopeptidase activity. Other putative targets include members of the Toll signalling pathway and proteins with kinase, ATPase, protease, scavenger receptor or Lectin C-type activity or that participate in fatty acid biosynthesis or oxidative stress. Our results suggest that several specific miRNAs help regulate the cellular functions that maintain equilibrium between viral replication and the antiviral response during persistent infection of mosquito cells. This study is the first report of a global miRNA profile in a mosquito cell line persistently infected with DENV.
Collapse
Affiliation(s)
- Rodolfo Gamaliel Avila-Bonilla
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Martha Yocupicio-Monroy
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290, Del Valle Sur, Mexico City, CP 03100, Mexico.
| | - Laurence A Marchat
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Mónica A De Nova-Ocampo
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| | - Rosa María Del Ángel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados-IPN. Av, Instituto Politécnico Nacional 2508, San Pedro Zacatenco, Mexico City, CP 07360, Mexico.
| | - Juan Santiago Salas-Benito
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico; Programa de Doctorado en Ciencias en Biotecnología, Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, Guillermo Massieu Helguera 249, La Escalera-Ticomán, Mexico City, CP 07320, Mexico.
| |
Collapse
|
78
|
Baxter RHG, Contet A, Krueger K. Arthropod Innate Immune Systems and Vector-Borne Diseases. Biochemistry 2017; 56:907-918. [PMID: 28072517 DOI: 10.1021/acs.biochem.6b00870] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Arthropods, especially ticks and mosquitoes, are the vectors for a number of parasitic and viral human diseases, including malaria, sleeping sickness, Dengue, and Zika, yet arthropods show tremendous individual variation in their capacity to transmit disease. A key factor in this capacity is the group of genetically encoded immune factors that counteract infection by the pathogen. Arthropod-specific pattern recognition receptors and protease cascades detect and respond to infection. Proteins such as antimicrobial peptides, thioester-containing proteins, and transglutaminases effect responses such as lysis, phagocytosis, melanization, and agglutination. Effector responses are initiated by damage signals such as reactive oxygen species signaling from epithelial cells and recognized by cell surface receptors on hemocytes. Antiviral immunity is primarily mediated by siRNA pathways but coupled with interferon-like signaling, antimicrobial peptides, and thioester-containing proteins. Molecular mechanisms of immunity are closely linked to related traits of longevity and fertility, and arthropods have the capacity for innate immunological memory. Advances in understanding vector immunity can be leveraged to develop novel control strategies for reducing the rate of transmission of both ancient and emerging threats to global health.
Collapse
Affiliation(s)
- Richard H G Baxter
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| | - Alicia Contet
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| | - Kathryn Krueger
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| |
Collapse
|
79
|
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: 98] [Impact Index Per Article: 12.3] [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
|
80
|
Biron D, Nedelkov D, Missé D, Holzmuller P. Proteomics and Host–Pathogen Interactions. GENETICS AND EVOLUTION OF INFECTIOUS DISEASES 2017. [PMCID: PMC7149668 DOI: 10.1016/b978-0-12-799942-5.00011-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
81
|
Saraiva RG, Kang S, Simões ML, Angleró-Rodríguez YI, Dimopoulos G. Mosquito gut antiparasitic and antiviral immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:53-64. [PMID: 26827888 DOI: 10.1016/j.dci.2016.01.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/16/2016] [Accepted: 01/26/2016] [Indexed: 06/05/2023]
Abstract
Mosquitoes are responsible for the transmission of diseases with a serious impact on global human health, such as malaria and dengue. All mosquito-transmitted pathogens complete part of their life cycle in the insect gut, where they are exposed to mosquito-encoded barriers and active factors that can limit their development. Here we present the current understanding of mosquito gut immunity against malaria parasites, filarial worms, and viruses such as dengue, Chikungunya, and West Nile. The most recently proposed immune mediators involved in intestinal defenses are discussed, as well as the synergies identified between the recognition of gut microbiota and the mounting of the immune response.
Collapse
Affiliation(s)
- Raúl G Saraiva
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Yesseinia I Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
82
|
Wichit S, Ferraris P, Choumet V, Missé D. The effects of mosquito saliva on dengue virus infectivity in humans. Curr Opin Virol 2016; 21:139-145. [PMID: 27770704 DOI: 10.1016/j.coviro.2016.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/14/2022]
Abstract
Arboviruses such as Dengue, Chikungunya, and Zika viruses represent a major public health problem due to globalization and propagation of susceptible vectors worldwide. Arthropod vector-derived salivary factors have the capacity to modulate human cells function by enhancing or suppressing viral replication and, therefore, modify the establishment of local and systemic viral infection. Here, we discuss how mosquito saliva may interfere with Dengue virus (DENV) infection in humans. Identification of saliva factors that enhance infectivity will allow the production of vector-based vaccines and therapeutics that would interfere with viral transmission by targeting arthropod saliva components. Understanding the role of salivary proteins in DENV transmission will provide tools to control not only Dengue but also other arboviral diseases transmitted by the same vectors.
Collapse
Affiliation(s)
| | - Pauline Ferraris
- Laboratory of MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France
| | - Valérie Choumet
- Environment and Infectious Risks Unit, Pasteur Institute, Paris, France
| | - Dorothée Missé
- Laboratory of MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France.
| |
Collapse
|
83
|
Muturi EJ, Bara JJ, Rooney AP, Hansen AK. Midgut fungal and bacterial microbiota of Aedes triseriatus and Aedes japonicus shift in response to La Crosse virus infection. Mol Ecol 2016; 25:4075-90. [PMID: 27357374 DOI: 10.1111/mec.13741] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/10/2016] [Accepted: 06/27/2016] [Indexed: 01/20/2023]
Abstract
Understanding how midgut microbial communities of field-collected mosquitoes interact with pathogens is critical for controlling vector infection and disease. We used 16S rRNA and internal transcribed spacer sequencing to characterize the midgut bacterial and fungal communities of adult females of Aedes triseriatus and Aedes japonicus collected as pupae in tree holes, plastic bins and waste tires and their response to La Crosse virus (LACV) infection. For both mosquito species and across all habitat and virus treatments, a total of 62 bacterial operational taxonomic units (OTUs) from six phyla and 21 fungal OTUs from two phyla were identified. The majority of bacterial (92%) and fungal (71%) OTUs were shared between the mosquito species; however, several OTUs were unique to each species. Bacterial and fungal communities of individuals that took either infectious or noninfectious bloodmeals were less diverse and more homogeneous compared to those of newly emerged adults. Interestingly, LACV-infected A. triseriatus and A. japonicus had higher bacterial richness and lower fungal richness compared to individuals that took a noninfectious bloodmeal, suggesting that viral infection was associated with an increase in bacterial OTUs and a decrease in fungal OTUs. For both mosquito species, several OTUs were identified that had both high fidelity and specificity to mosquito midguts that were infected with LACV. Overall, these findings demonstrate that bacterial and fungal communities that reside in mosquito midguts respond to host diet and viral infection and could play a role in modulating vector susceptibility to LACV.
Collapse
Affiliation(s)
- Ephantus J Muturi
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, 1816 S. Oak St., Champaign, IL, 61820, USA
| | - Jeffrey J Bara
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, 1816 S. Oak St., Champaign, IL, 61820, USA.,Department of Biology, University of Louisville, Louisville, KY, 40292, USA
| | - Alejandro P Rooney
- Crop Bioprotection Research Unit, USDA, ARS, 1815 N. University St., Peoria, IL, 61604, USA
| | - Allison K Hansen
- Department of Entomology, University of Illinois at Urbana-Champaign, 505 S. Goodwin Ave., Urbana, IL, 61801, USA
| |
Collapse
|
84
|
Li X, Meng K, Qiao J, Liu H, Zhong C, Liu Q. Identification of Aadnr1, a novel gene related to innate immunity and apoptosis in Aedes albopictus. Gene 2016; 587:18-26. [PMID: 27045774 DOI: 10.1016/j.gene.2016.03.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 12/01/2022]
Abstract
Innate immunity and apoptosis play critical roles in defending pathogens in insects. In Drosophila, Dnr1 was reported as a negative regulator of apoptosis and immune deficiency (Imd) pathway which belongs to innate immunity. Aedes albopictus is an important kind of arbovirus vector and becoming a significant threat to public health due to its rapid global expansion. Here we identified an ortholog of dnr1 from A. albopictus, named as Aadnr1. Aadnr1 encoded a putative protein containing an N-terminal FERM domain and a C-terminal RING domain. AaDnr1 shared high identity with dipteran insects Dnr1 orthologs. Phylogenetic analyses showed that the closest relative of AaDnr1 was Aedes aegypti Dnr1. Real-time PCR proved that Aadnr1 mRNA was expressed ubiquitously during developmental and adult stages. Transcriptional levels of Aadnr1 were decreased drastically in C6/36 cells underwent apoptosis induced by Actinomycin D (Act D) treatment. Partial silence of Aadnr1 enhanced Act D-induced caspase activity. When challenged by heat-inactivated E. coli, transcriptional level of Aadnr1 was also decreased dramatically in C6/36 cells. While when C6/36 cells were infected with Sindbis virus TE/GFP, transcriptional level of Aadnr1 was reduced and recovered repeatedly, with an overall decreasing trend. It was also shown in this study that similar to Drosophila Dnr1, RING domain destabilized AaDnr1 protein. Taken together, the study identified an innate immunity and apoptosis related gene Aadnr1 in A. albopictus.
Collapse
Affiliation(s)
- Xiaomei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Kun Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jialu Qiao
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Hao Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chunyan Zhong
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Qingzhen Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China.
| |
Collapse
|
85
|
Butt TM, Coates CJ, Dubovskiy IM, Ratcliffe NA. Entomopathogenic Fungi: New Insights into Host-Pathogen Interactions. ADVANCES IN GENETICS 2016; 94:307-64. [PMID: 27131329 DOI: 10.1016/bs.adgen.2016.01.006] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Although many insects successfully live in dangerous environments exposed to diverse communities of microbes, they are often exploited and killed by specialist pathogens. Studies of host-pathogen interactions (HPI) provide valuable insights into the dynamics of the highly aggressive coevolutionary arms race between entomopathogenic fungi (EPF) and their arthropod hosts. The host defenses are designed to exclude the pathogen or mitigate the damage inflicted while the pathogen responds with immune evasion and utilization of host resources. EPF neutralize their immediate surroundings on the insect integument and benefit from the physiochemical properties of the cuticle and its compounds that exclude competing microbes. EPF also exhibit adaptations aimed at minimizing trauma that can be deleterious to both host and pathogen (eg, melanization of hemolymph), form narrow penetration pegs that alleviate host dehydration and produce blastospores that lack immunogenic sugars/enzymes but facilitate rapid assimilation of hemolymph nutrients. In response, insects deploy an extensive armory of hemocytes and macromolecules, such as lectins and phenoloxidase, that repel, immobilize, and kill EPF. New evidence suggests that immune bioactives work synergistically (eg, lysozyme with antimicrobial peptides) to combat infections. Some proteins, including transferrin and apolipophorin III, also demonstrate multifunctional properties, participating in metabolism, homeostasis, and pathogen recognition. This review discusses the molecular intricacies of these HPI, highlighting the interplay between immunity, stress management, and metabolism. Increased knowledge in this area could enhance the efficacy of EPF, ensuring their future in integrated pest management programs.
Collapse
Affiliation(s)
- T M Butt
- Swansea University, Swansea, Wales, United Kingdom
| | - C J Coates
- Swansea University, Swansea, Wales, United Kingdom
| | | | - N A Ratcliffe
- Swansea University, Swansea, Wales, United Kingdom; Universidade Federal Fluminense, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
86
|
Sim S, Hibberd ML. Genomic approaches for understanding dengue: insights from the virus, vector, and host. Genome Biol 2016; 17:38. [PMID: 26931545 PMCID: PMC4774013 DOI: 10.1186/s13059-016-0907-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The incidence and geographic range of dengue have increased dramatically in recent decades. Climate change, rapid urbanization and increased global travel have facilitated the spread of both efficient mosquito vectors and the four dengue virus serotypes between population centers. At the same time, significant advances in genomics approaches have provided insights into host–pathogen interactions, immunogenetics, and viral evolution in both humans and mosquitoes. Here, we review these advances and the innovative treatment and control strategies that they are inspiring.
Collapse
Affiliation(s)
- Shuzhen Sim
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Martin L Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
87
|
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: 143] [Impact Index Per Article: 15.9] [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
|
88
|
Ryabov EV, Fannon JM, Moore JD, Wood GR, Evans DJ. The Iflaviruses Sacbrood virus and Deformed wing virus evoke different transcriptional responses in the honeybee which may facilitate their horizontal or vertical transmission. PeerJ 2016; 4:e1591. [PMID: 26819848 PMCID: PMC4727977 DOI: 10.7717/peerj.1591] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/21/2015] [Indexed: 11/24/2022] Open
Abstract
Sacbrood virus (SBV) and Deformed wing virus (DWV) are evolutionarily related positive-strand RNA viruses, members of the Iflavirus group. They both infect the honeybee Apis mellifera but have strikingly different levels of virulence when transmitted orally. Honeybee larvae orally infected with SBV usually accumulate high levels of the virus, which halts larval development and causes insect death. In contrast, oral DWV infection at the larval stage usually causes asymptomatic infection with low levels of the virus, although high doses of ingested DWV could lead to DWV replicating to high levels. We investigated effects of DWV and SBV infection on the transcriptome of honeybee larvae and pupae using global RNA-Seq and real-time PCR analysis. This showed that high levels of SBV replication resulted in down-regulation of the genes involved in cuticle and muscle development, together with changes in expression of putative immune-related genes. In particular, honeybee larvae with high levels of SBV replication, with and without high levels of DWV replication, showed concerted up-regulated expression of antimicrobial peptides (AMPs), and down-regulated expression of the prophenoloxidase activating enzyme (PPAE) together with up-regulation of the expression of a putative serpin, which could lead to the suppression of the melanisation pathway. The effects of high SBV levels on expression of these immune genes were unlikely to be a consequence of SBV-induced developmental changes, because similar effects were observed in honeybee pupae infected by injection. In the orally infected larvae with high levels of DWV replication alone we observed no changes of AMPs or of gene expression in the melanisation pathway. In the injected pupae, high levels of DWV alone did not alter expression of the tested melanisation pathway genes, but resulted in up-regulation of the AMPs, which could be attributed to the effect of DWV on the regulation of AMP expression in response to wounding. We propose that the difference in expression of the honeybee immune genes induced by SBV and DWV may be an evolutionary adaptation to the different predominant transmission routes used by these viruses.
Collapse
Affiliation(s)
- Eugene V. Ryabov
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Jessica M. Fannon
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Jonathan D. Moore
- Warwick Systems Biology Centre, University of Warwick, Coventry, United Kingdom
| | - Graham R. Wood
- Warwick Systems Biology Centre, University of Warwick, Coventry, United Kingdom
| | - David J. Evans
- Biomedical Sciences Research Complex, University of St. Andrews, St. Andrews, United Kingdom
| |
Collapse
|
89
|
Epand RM. Antiviral Host Defence Peptides. HOST DEFENSE PEPTIDES AND THEIR POTENTIAL AS THERAPEUTIC AGENTS 2016. [PMCID: PMC7123656 DOI: 10.1007/978-3-319-32949-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The ongoing global mortality and morbidity associated with viral pathogens highlights the need for the continued development of effective, novel antiviral molecules. The antiviral activity of cationic host defence peptides is of significant interest as novel therapeutics for treating viral infection and predominantly due to their broad spectrum antiviral activity. These peptides also display powerful immunomodulatory activity and are key mediators of inflammation. Therefore, they offer a significant opportunity to inform the development of novel therapeutics for treating viral infections by either directly targeting the pathogen or by enhancing the innate immune response. In this chapter, we review the antiviral activity of cathelicidins and defensins, and examine the potential for these peptides to be used as novel antiviral agents.
Collapse
Affiliation(s)
- Richard M. Epand
- Health Sciences Centre, McMaster University, Hamilton, Ontario Canada
| |
Collapse
|
90
|
Viral Interference and Persistence in Mosquito-Borne Flaviviruses. J Immunol Res 2015; 2015:873404. [PMID: 26583158 PMCID: PMC4637105 DOI: 10.1155/2015/873404] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/30/2022] Open
Abstract
Mosquito-borne flaviviruses are important pathogens for humans, and the detection of two or more flaviviruses cocirculating in the same geographic area has often been reported. However, the epidemiological impact remains to be determined. Mosquito-borne flaviviruses are primarily transmitted through Aedes and Culex mosquitoes; these viruses establish a life-long or persistent infection without apparent pathological effects. This establishment requires a balance between virus replication and the antiviral host response. Viral interference is a phenomenon whereby one virus inhibits the replication of other viruses, and this condition is frequently associated with persistent infections. Viral interference and persistent infection are determined by several factors, such as defective interfering particles, competition for cellular factors required for translation/replication, and the host antiviral response. The interaction between two flaviviruses typically results in viral interference, indicating that these viruses share common features during the replicative cycle in the vector. The potential mechanisms involved in these processes are reviewed here.
Collapse
|
91
|
A cecropin-like antimicrobial peptide with anti-inflammatory activity from the black fly salivary glands. Parasit Vectors 2015; 8:561. [PMID: 26497304 PMCID: PMC4620007 DOI: 10.1186/s13071-015-1176-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/20/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Several antimicrobial peptides (AMPs) belonging to the cecropin family have been identified from the salivary glands of different black fly species, however, the immunological functions for these molecules were poorly understood. METHODS A novel cecropin-like antimicrobial peptide (SibaCec) was purified using reverse phase high-performance liquid chromatography (RP-HPLC) from the salivary glands of the black fly Simulium bannaense. The amino acid sequence of SibaCec was determined by a combination method of automated Edman degradation and cDNA sequencing. The morphologic changes of Gram-negative bacteria Escherichia coli treated with SibaCec were assessed by scanning electron microscopy (SEM). Quantitative PCR (qPCR) was performed to analyze the mRNA expression of the inducible NO synthase (iNOS) and pro-inflammatory cytokines. Nitric oxide (NO) generation was examined using a Griess assay and the secretion of pro-inflammatory cytokines was determined by an enzyme-linked immunosorbent assay (ELISA). The activation of extracellular signal-regulated kinase (ERK), p38, and the nuclear translocation of nuclear factor-kappaB (NF-κB) were assessed by Western blotting analysis. Circular dichroism (CD) spectroscopy was performed to evaluate the secondary structure of SibaCec in solvent environment. Interaction of SibaCec with lipopolysaccharide (LPS) was studied using fluorescein isothiocyanate (FITC)- conjugated LPS aggregates. Neutralization of LPS by SibaCec was assayed with the chromogenic limulus amebocyte lysate (LAL) test. qPCR was also used to analyze the expression of SibaCec mRNA in the salivary glands of insects after oral infection with the bacteria E.coli. RESULTS SibaCec possessed potent antimicrobial activity against Gram-negative bacteria, and showed low cytotoxicity toward mammalian cells. SEM analysis indicated that SibaCec killed bacteria through the disruption of cell membrane integrity. Furthermore, SibaCec significantly inhibited lipopolysaccharide (LPS)-induced production of NO and pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interferon-1β (IL-1β) and interferon-6 (IL-6) by blocking the activation of MAPKs and NF-κB signaling pathways. It mainly adopted an α-helix conformation in membrane-mimetic environments. SibaCec could interact and neutralize LPS. Infection of black flies with bacteria caused an upregulation of the expression of SibaCec. CONCLUSIONS These results demonstrated that in addition to the bactericidal capacity, SibaCec can function as immune regulator, inhibiting host secretion of inflammatory factors.
Collapse
|
92
|
Bacterial Exposure at the Larval Stage Induced Sexual Immune Dimorphism and Priming in Adult Aedes aegypti Mosquitoes. PLoS One 2015; 10:e0133240. [PMID: 26181517 PMCID: PMC4504673 DOI: 10.1371/journal.pone.0133240] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/25/2015] [Indexed: 11/23/2022] Open
Abstract
Gender differences in the immune response of insects are driven by natural selection for females and sexual selection for males. These natural forces entail a multitude of extrinsic and intrinsic factors involved in a genotype-environment interaction that results in sex-biased expression of the genes shared by males and females. However, little is known about how an infection at a particular ontogenetic stage may influence later stages, or how it may impact sexual immune dimorphism. Using Aedes aegypti mosquitoes, the aim of the present study was to analyze the effect of a bacterial exposure at the larval stage on adult immunity in males and females. The parameters measured were phenoloxidase activity, nitric oxide production, antimicrobial activity, and the antimicrobial peptide transcript response. As a measure of the immune response success, the persistence of injected bacteria was also evaluated. The results show that males, as well as females, were able to enhance survival in the adult stage as a result of being exposed at the larval stage, which indicates a priming effect. Moreover, there was a differential gender immune response, evidenced by higher PO activity in males as well as higher NO production and greater antimicrobial activity in females. The greater bacterial persistence in females suggests a gender-specific strategy for protection after a previous experience with an elicitor. Hence, this study provides a primary characterization of the complex and gender-specific immune response of male and female adults against a bacterial challenge in mosquitoes primed at an early ontogenetic stage.
Collapse
|
93
|
Dieng H, Hassan RB, Hassan AA, Ghani IA, Abang FB, Satho T, Miake F, Ahmad H, Fukumitsu Y, Hashim NA, Zuharah WF, Kassim NFA, Majid AHA, Selvarajoo R, Nolasco-Hipolito C, Ajibola OO, Tuen AA. Occurrence of a mosquito vector in bird houses: Developmental consequences and potential epidemiological implications. Acta Trop 2015; 145:68-78. [PMID: 25617636 DOI: 10.1016/j.actatropica.2015.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/29/2014] [Accepted: 01/07/2015] [Indexed: 11/25/2022]
Abstract
Even with continuous vector control, dengue is still a growing threat to public health in Southeast Asia. Main causes comprise difficulties in identifying productive breeding sites and inappropriate targeted chemical interventions. In this region, rural families keep live birds in backyards and dengue mosquitoes have been reported in containers in the cages. To focus on this particular breeding site, we examined the capacity of bird fecal matter (BFM) from the spotted dove, to support Aedes albopictus larval growth. The impact of BFM larval uptake on some adult fitness traits influencing vectorial capacity was also investigated. In serial bioassays involving a high and low larval density (HD and LD), BFM and larval standard food (LSF) affected differently larval development. At HD, development was longer in the BFM environment. There were no appreciable mortality differences between the two treatments, which resulted in similar pupation and adult emergence successes. BFM treatment produced a better gender balance. There were comparable levels of blood uptake and egg production in BFM and LSF females at LD; that was not the case for the HD one, which resulted in bigger adults. BFM and LSF females displayed equivalent lifespans; in males, this parameter was shorter in those derived from the BFM/LD treatment. Taken together these results suggest that bird defecations successfully support the development of Ae. albopictus. Due to their cryptic aspects, containers used to supply water to encaged birds may not have been targeted by chemical interventions.
Collapse
|
94
|
Xiao X, Zhang R, Pang X, Liang G, Wang P, Cheng G. A neuron-specific antiviral mechanism prevents lethal flaviviral infection of mosquitoes. PLoS Pathog 2015; 11:e1004848. [PMID: 25915054 PMCID: PMC4411065 DOI: 10.1371/journal.ppat.1004848] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/01/2015] [Indexed: 11/24/2022] Open
Abstract
Mosquitoes are natural vectors for many etiologic agents of human viral diseases. Mosquito-borne flaviviruses can persistently infect the mosquito central nervous system without causing dramatic pathology or influencing the mosquito behavior and lifespan. The mechanism by which the mosquito nervous system resists flaviviral infection is still largely unknown. Here we report that an Aedes aegypti homologue of the neural factor Hikaru genki (AaHig) efficiently restricts flavivirus infection of the central nervous system. AaHig was predominantly expressed in the mosquito nervous system and localized to the plasma membrane of neural cells. Functional blockade of AaHig enhanced Dengue virus (DENV) and Japanese encephalitis virus (JEV), but not Sindbis virus (SINV), replication in mosquito heads and consequently caused neural apoptosis and a dramatic reduction in the mosquito lifespan. Consistently, delivery of recombinant AaHig to mosquitoes reduced viral infection. Furthermore, the membrane-localized AaHig directly interfaced with a highly conserved motif in the surface envelope proteins of DENV and JEV, and consequently interrupted endocytic viral entry into mosquito cells. Loss of either plasma membrane targeting or virion-binding ability rendered AaHig nonfunctional. Interestingly, Culex pipien pallens Hig also demonstrated a prominent anti-flavivirus activity, suggesting a functionally conserved function for Hig. Our results demonstrate that an evolutionarily conserved antiviral mechanism prevents lethal flaviviral infection of the central nervous system in mosquitoes, and thus may facilitate flaviviral transmission in nature. The central nervous system plays a predominant role in organisms associated with cognition and higher-order functions, which is key to their normal behavior and successful survival. Many mosquito-borne flaviviruses particularly invade the central nervous system in vertebrates, resulting in dramatic neural degeneration and damage. As natural vectors, mosquitoes are highly permissive to flaviviral infection that can be persistent in the mosquito nervous system. However, the infection intriguingly does neither lead to significant malignant pathological sequelae, nor dramatically influences mosquito behavior or lifespan, and thus mosquitoes can transmit viruses efficiently. Little is known about the neuron-specific resistant mechanism in viral infection of mosquitoes. Here we report that a neuron-specific factor specifically controls flaviviral replication in the mosquito nervous system by interfering with viral entry, and its activity prevents lethal flaviviral infection of mosquitoes. Our study provides insight into the sophisticated interactions between mosquito-borne viruses and their vectors, and offers an important target for arboviral limitation in nature.
Collapse
Affiliation(s)
- Xiaoping Xiao
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, People's Republic of China
| | - Rudian Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
- School of Life Science, Tsinghua University, Beijing, People's Republic of China
| | - Xiaojing Pang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Guodong Liang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, People's Republic of China
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Viral Disease Control and Prevention, Beijing, People's Republic of China
| | - Penghua Wang
- Department of Microbiology and Immunology, School of Medicine, New York Medical College, Valhalla, New York, United States of America
| | - Gong Cheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
95
|
Liu Y, Zhou Y, Wu J, Zheng P, Li Y, Zheng X, Puthiyakunnon S, Tu Z, Chen XG. The expression profile of Aedes albopictus miRNAs is altered by dengue virus serotype-2 infection. Cell Biosci 2015; 5:16. [PMID: 25922662 PMCID: PMC4411651 DOI: 10.1186/s13578-015-0009-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/01/2015] [Indexed: 12/21/2022] Open
Abstract
Background Aedes albopictus is an important vector of Dengue virus (DENV) and it has quickly invaded the tropical and temperate environments worldwide. A few studies have shown that, microRNAs (miRNAs) regulate mosquito defense against pathogens. However, there is no systematic analysis of the impact of DENV infection on miRNA expression in Ae. albopictus. We conducted this study to investigate the miRNA expression of Ae. albopictus upon DENV-2 infection using Illumina RNA sequencing. Results A total of 103 known and 5 novel candidate miRNAs were identified in DENV-2 infected and non-infected adult female Ae. albopictus. Comparative analysis indicated that 52 miRNAs were significantly down-regulated and 18 were up-regulated significantly after infection. Furthermore, RT-qPCR validated the expression patterns of eleven of these differentially expressed miRNAs. Targets prediction and functional analysis of these regulated miRNAs suggested that miR-34-5p and miR-87 might be involved in the anti-pathogen and immune responses. Conclusion This is the first systematic study on the impact of DENV infection on miRNA expression in Ae. albopictus. Complex changes in miRNA expression suggest a potential role of miRNAs in antiviral responses by regulating immune-related genes. This investigation provides information concerning DENV-induced miRNAs and offers clues for identifying potential candidates for vector based antiviral strategies. Electronic supplementary material The online version of this article (doi:10.1186/s13578-015-0009-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanxia Liu
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Yanhe Zhou
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Jinya Wu
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Peiming Zheng
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Yiji Li
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Xiaoying Zheng
- Department of Parasitology, School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong P.R. China
| | - Santhosh Puthiyakunnon
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| | - Zhijian Tu
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia USA
| | - Xiao-Guang Chen
- Key Laboratory of Prevention and Control for Emerging Infectious Diseases of Guangdong Higher Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong P.R. China
| |
Collapse
|
96
|
Kean J, Rainey SM, McFarlane M, Donald CL, Schnettler E, Kohl A, Pondeville E. Fighting Arbovirus Transmission: Natural and Engineered Control of Vector Competence in Aedes Mosquitoes. INSECTS 2015; 6:236-78. [PMID: 26463078 PMCID: PMC4553541 DOI: 10.3390/insects6010236] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/18/2015] [Accepted: 03/10/2015] [Indexed: 01/09/2023]
Abstract
Control of aedine mosquito vectors, either by mosquito population reduction or replacement with refractory mosquitoes, may play an essential role in the fight against arboviral diseases. In this review, we will focus on the development and application of biological approaches, both natural or engineered, to limit mosquito vector competence for arboviruses. The study of mosquito antiviral immunity has led to the identification of a number of host response mechanisms and proteins that are required to control arbovirus replication in mosquitoes, though more factors influencing vector competence are likely to be discovered. We will discuss key aspects of these pathways as targets either for selection of naturally resistant mosquito populations or for mosquito genetic manipulation. Moreover, we will consider the use of endosymbiotic bacteria such as Wolbachia, which in some cases have proven to be remarkably efficient in disrupting arbovirus transmission by mosquitoes, but also the use of naturally occurring insect-specific viruses that may interfere with arboviruses in mosquito vectors. Finally, we will discuss the use of paratransgenesis as well as entomopathogenic fungi, which are also proposed strategies to control vector competence.
Collapse
Affiliation(s)
- Joy Kean
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Stephanie M Rainey
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Melanie McFarlane
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Esther Schnettler
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Emilie Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| |
Collapse
|
97
|
Aida HN, Dieng H, Ahmad AH, Satho T, Nurita AT, Salmah MRC, Miake F, Norasmah B. The biology and demographic parameters of Aedes albopictus in northern peninsular Malaysia. Asian Pac J Trop Biomed 2015; 1:472-7. [PMID: 23569816 DOI: 10.1016/s2221-1691(11)60103-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 04/27/2011] [Accepted: 05/11/2011] [Indexed: 10/28/2022] Open
Abstract
OBJECTIVE To generate life table characteristics for the dengue vector Aedes albopictus (A. albopictus) under uncontrolled conditions, incorporating both the aquatic and the adult stages. METHODS Ten females derived from wild pupae were allowed to fully blood-feed on restrained mice. 774 eggs were hatched in seasoned water. F1 larvae were followed for development until their F2 counterparts emerged as adults. Some population parameters were monitored (F1) or estimated (F2). RESULTS A. albopictus exhibited increased fecundity and egg hatch success. Immature development was quick. Immature survival was high, with lowest rate in the pupal stage. Adult emergence was about 81% and sex ratio was close to 1:1. Generational mortality (K) was about 28%. A high proportion of females completed a reproductive cycle and the obtained parity rate was predicted to lead to higher fecundity in the next generation. CONCLUSIONS It can be concluded that natural A. albopictus populations in Penang seem largely determined by quick development in combination with low immature loss and increased oviposition.
Collapse
Affiliation(s)
- H Nur Aida
- Medical Entomology Laboratory, School of Biological Sciences, Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Marie A, Holzmuller P, Tchioffo MT, Rossignol M, Demettre E, Seveno M, Corbel V, Awono-Ambéné P, Morlais I, Remoue F, Cornelie S. Anopheles gambiae salivary protein expression modulated by wild Plasmodium falciparum infection: highlighting of new antigenic peptides as candidates of An. gambiae bites. Parasit Vectors 2014; 7:599. [PMID: 25526764 PMCID: PMC4287575 DOI: 10.1186/s13071-014-0599-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/10/2014] [Indexed: 12/24/2022] Open
Abstract
Background Malaria is the major parasitic disease worldwide caused by Plasmodium infection. The objective of integrated malaria control programs is to decrease malaria transmission, which needs specific tools to be accurately assessed. In areas where the transmission is low or has been substantially reduced, new complementary tools have to be developed to improve surveillance. A recent approach, based on the human antibody response to Anopheles salivary proteins, has been shown to be efficient in evaluating human exposure to Anopheles bites. The aim of the present study was to identify new An. gambiae salivary proteins as potential candidate biomarkers of human exposure to P. falciparum-infective bites. Methods Experimental infections of An. gambiae by wild P. falciparum were carried out in semi-field conditions. Then a proteomic approach, combining 2D-DIGE and mass spectrometry, was used to identify the overexpressed salivary proteins in infected salivary glands compared to uninfected An. gambiae controls. Subsequently, a peptide design of each potential candidate was performed in silico and their antigenicity was tested by an epitope-mapping technique using blood from individuals exposed to Anopheles bites. Results Five salivary proteins (gSG6, gSG1b, TRIO, SG5 and long form D7) were overexpressed in the infected salivary glands. Eighteen peptides were designed from these proteins and were found antigenic in children exposed to the Anopheles bites. Moreover, the results showed that the presence of wild P. falciparum in salivary glands modulates the expression of several salivary proteins and also appeared to induce post-translational modifications. Conclusions This study is, to our knowledge, the first that compares the sialome of An. gambiae both infected and not infected by wild P. falciparum, making it possible to mimic the natural conditions of infection. This is a first step toward a better understanding of the close interactions between the parasite and the salivary gland of mosquitoes. In addition, these results open the way to define biomarkers of infective bites of Anopheles, which could, in the future, improve the estimation of malaria transmission and the evaluation of malaria vector control tools. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0599-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra Marie
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Philippe Holzmuller
- CIRAD Département Systèmes Biologiques BIOS UMR 15 CMAEE "Contrôle des Maladies Exotiques et Emergentes", Campus International de Baillarguet, TA A-15/G, Montpellier cedex 5, 34398, France.
| | - Majoline T Tchioffo
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Marie Rossignol
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Edith Demettre
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, UM1, UM2, Plate-forme de Protéomique Fonctionnelle CNRS UMS BioCampus 3426, Montpellier, 34094, France.
| | - Martial Seveno
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, UM1, UM2, Plate-forme de Protéomique Fonctionnelle CNRS UMS BioCampus 3426, Montpellier, 34094, France.
| | - Vincent Corbel
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,Department of Entomology, Faculty of Agriculture, Kasetsart University, 50 Ngam Wong Wan Rd, Ladyaow Chatuchak, Bangkok, 10900, Thailand.
| | - Parfait Awono-Ambéné
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale (OCEAC), Yaoundé, BP 288, Cameroun.
| | - Isabelle Morlais
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale (OCEAC), Yaoundé, BP 288, Cameroun.
| | - Franck Remoue
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Sylvie Cornelie
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,MIVEGEC- IRD- CREC, Cotonou, 01 BP4414 RP, Bénin.
| |
Collapse
|
99
|
Galay RL, Umemiya-Shirafuji R, Mochizuki M, Fujisaki K, Tanaka T. Iron metabolism in hard ticks (Acari: Ixodidae): the antidote to their toxic diet. Parasitol Int 2014; 64:182-9. [PMID: 25527065 DOI: 10.1016/j.parint.2014.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 12/01/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022]
Abstract
Ticks are notorious parasitic arthropods, known for their completely host-blood-dependent lifestyle. Hard ticks (Acari: Ixodidae) feed on their hosts for several days and can ingest blood more than a hundred times their unfed weight. Their blood-feeding habit facilitates the transmission of various pathogens. It is remarkable how hard ticks cope with the toxic nature of their blood meal, which contains several molecules that can promote oxidative stress including iron. While it is required in several physiological processes, high amounts of iron can be dangerous because iron can also participate in the formation of free radicals that may cause cellular damage and death. Here we review the current knowledge on heme and inorganic iron metabolism in hard ticks and compare it with that in vertebrates and other arthropods. We briefly discuss the studies on heme transport, storage and detoxification, and the transport and storage of inorganic iron, with emphasis on the functions of tick ferritins. This review points out other aspects of tick iron metabolism that warrant further investigation, as compared to mammals and other arthropods. Further understanding of this physiological process may help in formulating new control strategies for tick infestation and the spread of tick-borne diseases.
Collapse
Affiliation(s)
- Remil Linggatong Galay
- Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan; Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Rika Umemiya-Shirafuji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Masami Mochizuki
- Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan; Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Kozo Fujisaki
- National Agricultural and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0856, Japan
| | - Tetsuya Tanaka
- Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi 753-8515, Japan; Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.
| |
Collapse
|
100
|
Bernard E, Hamel R, Neyret A, Ekchariyawat P, Molès JP, Simmons G, Chazal N, Desprès P, Missé D, Briant L. Human keratinocytes restrict chikungunya virus replication at a post-fusion step. Virology 2014; 476:1-10. [PMID: 25496825 DOI: 10.1016/j.virol.2014.11.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 11/06/2014] [Accepted: 11/12/2014] [Indexed: 11/26/2022]
Abstract
Transmission of chikungunya virus (CHIKV) to humans is initiated by puncture of the skin by a blood-feeding Aedes mosquito. Despite the growing knowledge accumulated on CHIKV, the interplay between skin cells and CHIKV following inoculation still remains unclear. In this study we questioned the behavior of human keratinocytes, the predominant cell population in the skin, following viral challenge. We report that CHIKV rapidly elicits an innate immune response in these cells leading to the enhanced transcription of type I/II and type III interferon genes. Concomitantly, we show that despite viral particles internalization into Rab5-positive endosomes and efficient fusion of virus and cell membranes, keratinocytes poorly replicate CHIKV as attested by absence of nonstructural proteins and genomic RNA synthesis. Accordingly, human keratinocytes behave as an antiviral defense against CHIKV infection rather than as a primary targets for initial replication. This picture significantly differs from that reported for Dengue and West Nile mosquito-borne viruses.
Collapse
Affiliation(s)
- Eric Bernard
- Centre d׳étude d'agents Pathogènes et Biotechnologies pour la Santé, CPBS CNRS- UMR5236/UM1/UM2, Montpellier, France
| | - Rodolphe Hamel
- Laboratoire Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution, Contrôle, UMR 5290 CNRS/IRD/UM1, Montpellier, France
| | - Aymeric Neyret
- Centre d׳étude d'agents Pathogènes et Biotechnologies pour la Santé, CPBS CNRS- UMR5236/UM1/UM2, Montpellier, France
| | - Peeraya Ekchariyawat
- Laboratoire Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution, Contrôle, UMR 5290 CNRS/IRD/UM1, Montpellier, France
| | | | - Graham Simmons
- Blood Systems Research Institute, San Francisco, CA 94118, USA
| | - Nathalie Chazal
- Centre d׳étude d'agents Pathogènes et Biotechnologies pour la Santé, CPBS CNRS- UMR5236/UM1/UM2, Montpellier, France
| | - Philippe Desprès
- Unité Interactions Moléculaires Flavivirus-Hôtes, Institut Pasteur, Paris, France
| | - Dorothée Missé
- Laboratoire Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution, Contrôle, UMR 5290 CNRS/IRD/UM1, Montpellier, France
| | - Laurence Briant
- Centre d׳étude d'agents Pathogènes et Biotechnologies pour la Santé, CPBS CNRS- UMR5236/UM1/UM2, Montpellier, France.
| |
Collapse
|