1
|
Wang Y, Ling L, Jiang L, Marin-Lopez A. Research progress toward arthropod salivary protein vaccine development for vector-borne infectious diseases. PLoS Negl Trop Dis 2024; 18:e0012618. [PMID: 39636798 PMCID: PMC11620354 DOI: 10.1371/journal.pntd.0012618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Hematophagous arthropods, including mosquitoes, ticks, and flies, are responsible for the transmission of several pathogens to vertebrates on whom they blood feed. The diseases caused by these pathogens, collectively known as vector-borne diseases (VBDs), threaten the health of humans and animals. In general, attempts to develop vaccines for pathogens transmitted by arthropods have met with moderate success, with few vaccine candidates currently developed. Nowadays, there are vaccine candidates under clinical trials, including different platforms, like mRNA, DNA, recombinant viral vector-based, virus-like particles (VLPs), inactivated-virus, live-attenuated virus, peptide and protein-based vaccines, all of them based on the presentation of pathogen antigens to the host immune system. A new approach to prevent VBDs has arose during the last decades, based on the design of vaccines that target vector-derived antigens. The salivary secretions of arthropods, in addition of causing allergic reactions and harbor pathogens, are also involved in the transmission and infection establishment in the host, altering its immune responses. In this review, we summarize the achievements in the arthropod salivary-based vaccine development for different vector-borne infectious diseases. This provides a rationale for creating vaccines against different types of arthropod salivary proteins, such as mosquitoes, ticks, and sand flies. Using salivary proteins of clinically important vectors might contribute to achieve protection against and control multiple arthropod-borne infection diseases.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Lin Ling
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Lijie Jiang
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
2
|
Zhang B, Chen L, Xiao S, Dang C, Wang F, Fang Q, Ye X, Stanley DW, Ye G. iSalivaomicDB: A comprehensive saliva omics database for insects. INSECT SCIENCE 2024; 31:1998-2001. [PMID: 38450904 DOI: 10.1111/1744-7917.13349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Longfei Chen
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Shan Xiao
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Cong Dang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Fang Wang
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Qi Fang
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Xinhai Ye
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - David W Stanley
- Biological Control of Insects Research Laboratory USDA/Agricultural Research Service, Columbia MO, USA
| | - Gongyin Ye
- State Key Laboratory of Rice Biology and Breeding, Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests & Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Yuan C, Xu Q, Ning Y, Xia Q. Potential mechanisms implied in tick infection by arboviruses and their transmission to vertebrate hosts. Integr Zool 2024. [PMID: 39016029 DOI: 10.1111/1749-4877.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Ticks can transmit many pathogens, including arboviruses, to their vertebrate hosts. Arboviruses must overcome or evade defense mechanisms during their passage from the tick gut to the hemolymph, salivary glands, and the feeding site in the host skin. This review summarizes current knowledge of defense mechanisms in specific tick tissues and at the feeding site in the host skin. We discuss the possible roles of these defense mechanisms in viral infection and transmission. The responses of tick salivary proteins to arbovirus infection are also discussed. This review provides information that may help accelerate research on virus-tick interactions.
Collapse
Affiliation(s)
- Chuanfei Yuan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qiong Xu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
| | - Yunjia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Qianfeng Xia
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
| |
Collapse
|
4
|
Wang ZY, Nie KX, Niu JC, Cheng G. Research progress toward the influence of mosquito salivary proteins on the transmission of mosquito-borne viruses. INSECT SCIENCE 2024; 31:663-673. [PMID: 37017683 DOI: 10.1111/1744-7917.13193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Mosquito-borne viruses (MBVs) are a large class of viruses transmitted mainly through mosquito bites, including dengue virus, Zika virus, Japanese encephalitis virus, West Nile virus, and chikungunya virus, which pose a major threat to the health of people around the world. With global warming and extended human activities, the incidence of many MBVs has increased significantly. Mosquito saliva contains a variety of bioactive protein components. These not only enable blood feeding but also play a crucial role in regulating local infection at the bite site and the remote dissemination of MBVs as well as in remodeling the innate and adaptive immune responses of host vertebrates. Here, we review the physiological functions of mosquito salivary proteins (MSPs) in detail, the influence and the underlying mechanism of MSPs on the transmission of MBVs, and the current progress and issues that urgently need to be addressed in the research and development of MSP-based MBV transmission blocking vaccines.
Collapse
Affiliation(s)
- Zhao-Yang Wang
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Kai-Xiao Nie
- Department of Pathogen Biology, School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ji-Chen Niu
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Gong Cheng
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Thambi PJ, Modahl CM, Kini RM. Niemann-Pick Type C2 Proteins in Aedes aegypti: Molecular Modelling and Prediction of Their Structure-Function Relationships. Int J Mol Sci 2024; 25:1684. [PMID: 38338961 PMCID: PMC10855982 DOI: 10.3390/ijms25031684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Aedes aegypti is a major vector that transmits arboviruses through the saliva injected into the host. Salivary proteins help in uninterrupted blood intake and enhance the transmission of pathogens. We studied Niemann-Pick Type C2 (NPC2) proteins, a superfamily of saliva proteins that play an important role in arbovirus infections. In vertebrates, a single conserved gene encodes for the NPC2 protein that functions in cholesterol trafficking. Arthropods, in contrast, have several genes that encode divergent NPC2 proteins. We compared the sequences of 20 A. aegypti NPC2 proteins to the cholesterol-binding residues of human and bovine, and fatty-acid-binding residues of ant NPC2 protein. We identified four mosquito NPC2 proteins as potential sterol-binding proteins. Two of these proteins (AAEL006854 and/or AAEL020314) may play a key role in ecdysteroid biosynthesis and moulting. We also identified one mosquito NPC2 protein as a potential fatty-acid-binding protein. Through molecular modelling, we predicted the structures of the potential sterol- and fatty-acid-binding proteins and compared them to the reference proteins.
Collapse
Affiliation(s)
| | - Cassandra M. Modahl
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
- Vector Biology Department, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - R. Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Department of Biochemistry and Molecular Biology, VCU School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
6
|
Mu X, Lin Z, Sun Y, Chen L, Lv Q, Ji C, Kuang X, Li W, Shang Z, Cheng J, Nie Y, Li Z, Wu J. Aedes albopictus salivary adenosine deaminase is an immunomodulatory factor facilitating dengue virus replication. Sci Rep 2023; 13:16660. [PMID: 37794048 PMCID: PMC10551004 DOI: 10.1038/s41598-023-43751-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023] Open
Abstract
The Asian tiger mosquito, Aedes albopictus, is an important vector for the transmission of arboviruses such as dengue virus (DENV). Adenosine deaminase (ADA) is a well-characterized metabolic enzyme involved in facilitating blood feeding and (or) arbovirus transmission in some hematophagous insect species. We previously reported the immunologic function of ADA by investigating its effect on mast cell activation and the interaction with mast cell tryptase and chymase. The 2-D gel electrophoresis and mass spectrometry analysis in the current study revealed that ADA is present and upregulated following mosquito blood feeding, as confirmed by qRT-PCR and western blot. In addition, the recombinant ADA efficiently converted adenosine to inosine. Challenging the Raw264.7 and THP-1 cells with recombinant ADA resulted in the upregulation of IL-1β, IL-6, TNF-α, CCL2, IFN-β, and ISG15. The current study further identified recombinant ADA as a positive regulator in NF-κB signaling targeting TAK1. It was also found that recombinant Ae. albopictus ADA facilitates the replication of DENV-2. Compared with cells infected by DENV-2 alone, the co-incubation of recombinant ADA with DENV-2 substantially increased IL-1β, IL-6, TNF-α, and CCL2 gene transcripts in Raw264.7 and THP-1 cells. However, the expression of IFN-β and ISG15 were markedly downregulated in Raw264.7 cells but upregulated in THP-1 cells. These findings suggest that the immunomodulatory protein, Ae. albopictus ADA is involved in mosquito blood feeding and may modulate DENV transmission via macrophage or monocyte-driven immune response.
Collapse
Affiliation(s)
- Xiaohui Mu
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
- Department of Reproductive Medicine, People's Hospital of Anshun City Guizhou Province, Anshun, 561000, Guizhou, China
| | - Zimin Lin
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yu Sun
- The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Lu Chen
- The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Qingqiao Lv
- Xi'an Peihua University, Xi'an, 710065, Shaanxi, China
| | - Cejuan Ji
- Department of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, Guizhou, China
| | - Xiaoyuan Kuang
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Weiyi Li
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Zhengling Shang
- Department of Immunology, College of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - Jinzhi Cheng
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Ying Nie
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Zhiqiang Li
- Department of Immunology, College of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China.
| | - Jiahong Wu
- Department of Parasitology, Provincial Key Laboratory of Modern Pathogen Biology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
7
|
Sri-In C, Thontiravong A, Bartholomay LC, Wechtaisong W, Thongmeesee K, Riana E, Tiawsirisup S. 34-kDa salivary protein enhances duck Tembusu virus infectivity in the salivary glands of Aedes albopictus by modulating the innate immune response. Sci Rep 2023; 13:9098. [PMID: 37277542 DOI: 10.1038/s41598-023-35914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023] Open
Abstract
Duck Tembusu virus (DTMUV) is an important flavivirus that can be transmitted to poultry via Aedes albopictus bites. Furthermore, humans residing in the DTMUV epidemic area display activated antiviral immune responses to local DTMUV isolates during the pathogenic invasion, thereby raising the primary concern that this flavivirus may be transmitted to humans via mosquito bites. Therefore, we identified the gene AALF004421, which is a homolog of the 34-kDa salivary protein (34 kDa) of Ae. albopictus and studied the salivary protein-mediated enhancement of DTMUV infection in Ae. albopictus salivary glands. We observed that double-stranded RNA-mediated silencing of the 34 kDa in mosquito salivary glands demonstrated that the silenced 34 kDa impaired DTMUV infectivity, similar to inhibition through serine protease. This impairment occurred as a consequence of triggering the innate immune response function of a macroglobulin complement-related factor (MCR). 34-kDa in the salivary gland which had similar activity as a serine protease, results in the abrogation of antimicrobial peptides production and strong enhance DTMUV replication and transmission. Although the function of the 34 kDa in Ae. albopictus is currently unknown; in the present study, we showed that it may have a major role in DTMUV infection in mosquito salivary glands through the suppression of the antiviral immune response in the earliest stages of infection. This finding provides the first identification of a prominently expressed 34 kDa protein in Ae. albopictus saliva that could serve as a target for controlling DTMUV replication in mosquito vectors.
Collapse
Affiliation(s)
- Chalida Sri-In
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Aunyaratana Thontiravong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin, USA
| | - Wittawat Wechtaisong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kritsada Thongmeesee
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Elizabeth Riana
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sonthaya Tiawsirisup
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
8
|
Lin DCD, Weng SC, Tsao PN, Chu JJH, Shiao SH. Co-infection of dengue and Zika viruses mutually enhances viral replication in the mosquito Aedes aegypti. Parasit Vectors 2023; 16:160. [PMID: 37165438 PMCID: PMC10172068 DOI: 10.1186/s13071-023-05778-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/16/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND The mosquito Aedes aegypti transmits two of the most serious mosquito-borne viruses, dengue virus (DENV) and Zika virus (ZIKV), which results in significant human morbidity and mortality worldwide. The quickly shifting landscapes of DENV and ZIKV endemicity worldwide raise concerns that their co-circulation through the Ae. aegypti mosquito vector could greatly exacerbate the disease burden in humans. Recent reports have indicated an increase in the number of co-infection cases in expanding co-endemic regions; however, the impact of co-infection on viral infection and the detailed molecular mechanisms remain to be defined. METHODS C6/36 (Aedes albopictus) cells were cultured in Dulbecco's modified Eagle medium/Mitsuhashi and Maramorosch Insect Medium (DMEM/MM) (1:1) containing 2% heat-inactivated fetal bovine serum and 1× penicillin/streptomycin solution. For virus propagation, the cells were infected with either DENV serotype 2 (DENV2) strain 16681 or ZIKV isolate Thailand/1610acTw (MF692778.1). Mosquitoes (Ae. aegypti UGAL [University of Georgia Laboratory]/Rockefeller strain) were orally infected with DENV2 and ZIKV through infectious blood-feeding. RESULTS We first examined viral replication activity in cells infected simultaneously, or sequentially, with DENV and ZIKV, and found interspecies binding of viral genomic transcripts to the non-structural protein 5 (NS5). When we challenged Ae. aegypti mosquitos with both DENV2 and ZIKV sequentially to probe similar interactions, virus production and vector susceptibility to infection were significantly enhanced. CONCLUSIONS Our results suggest that DENV2 and ZIKV simultaneously establishing infection in the Ae. aegypti mosquito vector may augment one another during replication. The data also implicate the homologous NS5 protein as a key intersection between the flaviviruses in co-infection, highlighting it as a potential target for vector control.
Collapse
Affiliation(s)
- Daniel Chieh-Ding Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
- Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
9
|
Marín-López A, Raduwan H, Chen TY, Utrilla-Trigo S, Wolfhard DP, Fikrig E. Mosquito Salivary Proteins and Arbovirus Infection: From Viral Enhancers to Potential Targets for Vaccines. Pathogens 2023; 12:371. [PMID: 36986293 PMCID: PMC10054260 DOI: 10.3390/pathogens12030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Arthropod-borne viruses present important public health challenges worldwide. Viruses such as DENV, ZIKV, and WNV are of current concern due to an increasing incidence and an expanding geographic range, generating explosive outbreaks even in non-endemic areas. The clinical signs associated with infection from these arboviruses are often inapparent, mild, or nonspecific, but occasionally develop into serious complications marked by rapid onset, tremors, paralysis, hemorrhagic fever, neurological alterations, or death. They are predominately transmitted to humans through mosquito bite, during which saliva is inoculated into the skin to facilitate blood feeding. A new approach to prevent arboviral diseases has been proposed by the observation that arthropod saliva facilitates transmission of pathogens. Viruses released within mosquito saliva may more easily initiate host invasion by taking advantage of the host's innate and adaptive immune responses to saliva. This provides a rationale for creating vaccines against mosquito salivary proteins, especially because of the lack of licensed vaccines against most of these viruses. This review aims to provide an overview of the effects on the host immune response by the mosquito salivary proteins and how these phenomena alter the infection outcome for different arboviruses, recent attempts to generate mosquito salivary-based vaccines against flavivirus including DENV, ZIKV, and WNV, and the potential benefits and pitfalls that this strategy involves.
Collapse
Affiliation(s)
- Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Sergio Utrilla-Trigo
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
- Center for Animal Health Research (CISA-INIA/CSIC), 28130 Madrid, Spain
| | - David P. Wolfhard
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
- Faculty of Engineering Sciences, Institute of Pharmacy and Molecular Biotechnology, 69120 Heidelberg, Germany
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
10
|
Bhardwaj A, Sharma R, Grover A. Immuno-informatics guided designing of a multi-epitope vaccine against Dengue and Zika. J Biomol Struct Dyn 2023; 41:1-15. [PMID: 34796791 DOI: 10.1080/07391102.2021.2002720] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Dengue and zika are amongst the most prevalent mosquito-borne diseases caused by closely related members Dengue virus (DENV) and Zika virus (ZIKV), respectively, of the Flaviviridae family. DENV and ZIKV have been reported to co-infect several people, resulting in fatalities across the world. A vaccine that can safeguard against both these pathogens concurrently, can offer several advantages. This study has employed immuno-informatics for devising a multi-epitope, multi-pathogenic vaccine against both these viruses. Since, the two viruses share a common vector source, whose salivary components are reported to aid viral pathogenesis; antigenic salivary proteins from Aedes aegypti were also incorporated into the design of the vaccine along with conserved structural and non-structural viral proteins. Conserved B- and T-cell epitopes were identified for all the selected antigenic proteins. These epitopes were merged and further supplemented with β-defensin as an adjuvant, to yield an immunogenic vaccine construct. In-silico 3D modeling and structural validation of the vaccine construct was conducted, followed by its molecular docking and molecular dynamics simulation studies with human TLR2. Immune simulation study was also performed, and it further provided support that the designed vaccine can mount an effective immune response and hence provide protection against both DENV and ZIKV. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aditi Bhardwaj
- School of Biosciences & Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Ritika Sharma
- School of Biotechnology, Jawaharlal Nehru University (JNU), Delhi, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University (JNU), Delhi, India
| |
Collapse
|
11
|
Arnoldi I, Mancini G, Fumagalli M, Gastaldi D, D'Andrea L, Bandi C, Di Venere M, Iadarola P, Forneris F, Gabrieli P. A salivary factor binds a cuticular protein and modulates biting by inducing morphological changes in the mosquito labrum. Curr Biol 2022; 32:3493-3504.e11. [PMID: 35835123 DOI: 10.1016/j.cub.2022.06.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/23/2022] [Accepted: 06/15/2022] [Indexed: 01/03/2023]
Abstract
The mosquito proboscis is an efficient microelectromechanical system, which allows the insect to feed on vertebrate blood quickly and painlessly. Its efficiency is further enhanced by the insect saliva, although through unclear mechanisms. Here, we describe the initial trigger of an unprecedented feedback signaling pathway in Aedes mosquitoes affecting feeding behavior. We identified LIPS proteins in the saliva of Aedes mosquitoes that promote feeding in the vertebrate skin. LIPS show a new all-helical protein fold constituted by two domains. The N-terminal domain interacts with a cuticular protein (Cp19) located at the tip of the mosquito labrum. Upon interaction, the morphology of the labral cuticle changes, and this modification is most likely sensed by proprioceptive neurons. Our study identifies an additional role of mosquito saliva and underlines that the external cuticle is a possible site of key molecular interactions affecting the insect biology and its vector competence.
Collapse
Affiliation(s)
- Irene Arnoldi
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; Entopar lab, Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy; Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy
| | - Giulia Mancini
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy
| | - Marco Fumagalli
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; Biochemistry Unit, Department Biology and Biotechnology, University of Pavia, Via Taramelli 3, 27100 Pavia, Italy
| | - Dario Gastaldi
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering Giulio Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | - Luca D'Andrea
- Laboratory of Biological Structure Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering Giulio Natta, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133, Milano, Italy
| | - Claudio Bandi
- Entopar lab, Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy; Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy
| | - Monica Di Venere
- Biochemistry Unit, Department Biology and Biotechnology, University of Pavia, Via Taramelli 3, 27100 Pavia, Italy
| | - Paolo Iadarola
- Biochemistry Unit, Department Biology and Biotechnology, University of Pavia, Via Taramelli 3, 27100 Pavia, Italy
| | - Federico Forneris
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy.
| | - Paolo Gabrieli
- The Armenise-Harvard Laboratory of Structural Biology, Department Biology and Biotechnology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; Entopar lab, Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy; Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy.
| |
Collapse
|
12
|
Weng SC, Shiao SH. SUMOylation Is Essential for Dengue Virus Replication and Transmission in the Mosquito Aedes aegypti. Front Microbiol 2022; 13:801284. [PMID: 35572621 PMCID: PMC9093690 DOI: 10.3389/fmicb.2022.801284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
Small ubiquitin-like modifier (SUMO) is a reversible post-translational protein modifier. Protein SUMOylation regulates a wide variety of cellular processes and is important for controlling virus replication. Earlier studies suggest that dengue virus envelope protein interacts with Ubc9, the sole E2-conjugating enzyme required for protein SUMOylation in mammalian cells. However, little is known about the effect of protein SUMOylation on dengue virus replication in the major dengue vector, Aedes aegypti. Thus, in this study, we investigated the impact of protein SUMOylation on dengue virus replication in A. aegypti. The transcription of A. aegypti Ubc9 was significantly increased in the midgut after a normal blood meal. Silencing AaUbc9 resulted in significant inhibition of dengue virus NS1 protein production, viral genome transcription, and reduced viral titer in the mosquito saliva. In addition, we showed that dengue virus E proteins and prM proteins were SUMOylated post-infection. The amino acid residues K51 and K241 of dengue virus E protein were essential for protein SUMOylation. Taken together, our results reveal that protein SUMOylation contributes to dengue virus replication and transmission in the mosquito A. aegypti. This study introduces the possibility that protein SUMOylation is beneficial for virus replication and facilitates virus transmission from the mosquito.
Collapse
Affiliation(s)
- Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
13
|
Sri-In C, Thontiravong A, Bartholomay LC, Tiawsirisup S. Effects of Aedes aegypti salivary protein on duck Tembusu virus replication and transmission in salivary glands. Acta Trop 2022; 228:106310. [PMID: 35032469 DOI: 10.1016/j.actatropica.2022.106310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/29/2022]
Abstract
Duck Tembusu virus (DTMUV) infection is an arthropod-borne viral disease that affects many poultry species, including ducks, chickens, and geese. Aedes aegypti mosquito is an important vector of DTMUV. This study sought to determine whether any individual Ae. aegypti salivary protein modulated DTMUV replication in the mosquito salivary gland. Ae. aegypti salivary gland protein of 34 kDa (AaSG34) was found to be expressed explicitly in mosquito salivary glands and was upregulated following DTMUV infection. Thus, AaSG34 was silenced in mosquitoes via RNA interference using double strand RNA (dsRNA), and the mosquitoes were then infected with DTMUV to elucidate their effects on DTMUV replication and transmission. Transcripts of the DTMUV genome in salivary glands and virus titer in saliva were significantly diminished when AaSG34 was silenced, indicating that its presence enhances DTMUV replication in the salivary glands and DTMUV dissemination to saliva. Furthermore, the expression of antimicrobial peptides (AMPs) was upregulated upon AaSG34 silenced. Our results demonstrate that AaSG34 may play a vital role in the suppression of antiviral immune responses to enhance DTMUV replication and transmission. We thus provide new information on the effect of the AaSG34 salivary protein on DTMUV replication in Ae. aegypti as the mechanism of blocking virus transmission to the host.
Collapse
Affiliation(s)
- Chalida Sri-In
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Aunyaratana Thontiravong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin, United States
| | - Sonthaya Tiawsirisup
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
14
|
Cheang KW, Chen WY, Wu-Hsieh BA, Shiao SH. Infecting mosquitoes alters DENV-2 characteristics and enhances hemorrhage-induction potential in Stat1-/- mice. PLoS Negl Trop Dis 2021; 15:e0009728. [PMID: 34449772 PMCID: PMC8428656 DOI: 10.1371/journal.pntd.0009728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 09/09/2021] [Accepted: 08/14/2021] [Indexed: 11/19/2022] Open
Abstract
Dengue is one of the most prevalent arthropod-borne viral diseases in humans. There is still no effective vaccine or treatment to date. Previous studies showed that mosquito-derived factors present in saliva or salivary gland extract (SGE) contribute to the pathogenesis of dengue. In this study, we aimed to investigate the interplay between mosquito vector and DENV and to address the question of whether the mosquito vector alters the virus that leads to consequential disease manifestations in the mammalian host. DENV2 cultured in C6/36 cell line (culture-DENV2) was injected to Aedes aegypti intrathoracically. Saliva was collected from infected mosquitoes 7 days later. Exploiting the sensitivity of Stat1-/- mice to low dose of DENV2 delivered intradermally, we showed that DENV2 collected in infected mosquito saliva (msq-DENV2) induced more severe hemorrhage in mice than their culture counterpart. Msq-DENV2 was characterized by smaller particle size, larger plaque size and more rapid growth in mosquito as well as mammalian cell lines compared to culture-DENV2. In addition, msq-DENV2 was more efficient than culture-DENV2 in inducing Tnf mRNA production by mouse macrophage. Together, our results point to the possibility that the mosquito vector provides an environment that alters DENV2 by changing its growth characteristics as well as its potential to cause disease.
Collapse
Affiliation(s)
- Ka Wan Cheang
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Yu Chen
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Betty A. Wu-Hsieh
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
- * E-mail: (BAW-H); (S-HS)
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan
- * E-mail: (BAW-H); (S-HS)
| |
Collapse
|
15
|
Weng SC, Tsao PN, Shiao SH. Blood glucose promotes dengue virus infection in the mosquito Aedes aegypti. Parasit Vectors 2021; 14:376. [PMID: 34311776 PMCID: PMC8314564 DOI: 10.1186/s13071-021-04877-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 11/30/2022] Open
Abstract
Background Dengue fever is the most rapidly spreading mosquito-borne viral disease globally. More than 2.5 billion people live in dengue-endemic areas. Previous studies suggested an interrelationship between diabetes mellitus (DM) and dengue hemorrhagic fever (DHF). Conversely, glycolysis is a critical metabolic pathway for optimal dengue virus (DENV) replication. However, little is known concerning the effect of glucose on DENV replication in mosquitoes. In this study, we investigated the impact of glucose on DENV replication in mosquitoes Aedes aegypti. Methods Mosquitoes (Ae. aegypti UGAL/Rockefeller strain) were orally infected with DENV (serotype 2, 16681 strain) through infectious blood feeding. The DENV infection and transmission rates were determined by examining mosquito bodies and saliva, respectively, for DENV positivity at different time points after infection. In addition, a reverse genetic approach was applied by introducing double-stranded RNA against genes of interest into the mosquitoes to inhibit gene expression. Results Our data revealed a significant increase of DENV genome levels in mosquitoes consuming an infectious blood meal supplemented with glucose, suggesting that blood glucose is an important factor for viral replication. Interestingly, a significant increase of DENV E protein levels was detected in the saliva 4 days faster in mosquitoes that consumed infectious blood meals supplemented with glucose than in those consuming infectious blood meals alone. Furthermore, we perform RNAi to silence AKT or TOR and investigate the molecular mechanism regulating the glucose-mediated enhancement of viral replication. Silencing of AKT or TOR significantly reduced DENV titers in mosquitoes. Conclusions This study suggested that blood glucose is beneficial to DENV replication and that it facilitates virus transmission in mosquitoes via AKT and TOR signaling. Therefore, our results strengthen our understanding of dengue fever and DM co-morbidity and possibly reveal new targets for specific antiviral therapies. Graphical abstract ![]()
Collapse
Affiliation(s)
- Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
16
|
Weng SC, Li HH, Li JC, Liu WL, Chen CH, Shiao SH. A Thioester-Containing Protein Controls Dengue Virus Infection in Aedes aegypti Through Modulating Immune Response. Front Immunol 2021; 12:670122. [PMID: 34054842 PMCID: PMC8155531 DOI: 10.3389/fimmu.2021.670122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023] Open
Abstract
Complement-like proteins in arthropods defend against invading pathogens in the early phases of infection. Thioester-containing proteins (TEPs), which exhibit high similarity to mammalian complement C3, are thought to play a key role in the innate immunity of arthropods. We identified and characterized anti-dengue virus (DENV) host factors, in particular complement-like proteins, in the mosquito Aedes aegypti. Our results indicate that TEP1 limits DENV infection in Ae. aegypti. We showed that TEP1 transcription is highly induced in mosquitoes following DENV infection. Silencing TEP1 resulted in the up-regulation of viral RNA and proteins. In addition, the production of infectious virus particles increased in the absence of TEP1. We generated a transgenic mosquito line with a TEP1 loss-of-function phenotype under a blood meal-inducible promoter. We showed that viral protein and titers increased in transgenic mosquitoes after an infectious blood meal. Interestingly, expression of transcription factor Rel2 and certain anti-microbial peptides (AMPs) were inhibited in transgenic mosquitoes. Overall, our results suggest that TEP1 regulates the immune response and consequently controls the replication of dengue virus in mosquitoes. This finding provides new insight into the molecular mechanisms of mosquito host factors in the regulation of DENV replication.
Collapse
Affiliation(s)
- Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsing-Han Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Wei-Liang Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Assis JB, Cogliati B, Esteves E, Capurro ML, Fonseca DM, Sá-Nunes A. Aedes aegypti mosquito saliva ameliorates acetaminophen-induced liver injury in mice. PLoS One 2021; 16:e0245788. [PMID: 33556084 PMCID: PMC7869984 DOI: 10.1371/journal.pone.0245788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) overdose is the most common cause of drug-induced liver injury (DILI). Although the primary hepatic damage is induced by APAP-derived toxic intermediates resulting from cytochrome P450 metabolism, immune components also play an important role in DILI pathophysiology. Aedes aegypti saliva is a source of bioactive molecules with in vitro anti-inflammatory and immunomodulatory activities. However, evidences on the therapeutic use of Ae. aegypti salivary preparations in animal models of relevant clinical conditions are still scarce. Thus, the present study was designed to evaluate the protective role of Ae. aegypti saliva in a murine model of APAP-induced DILI. C57BL/6 mice were exposed to Ae. aegypti bites 2 hours after APAP overdose. Biochemical and immunological parameters were evaluated in blood and liver samples at different time points after APAP administration. Exposure to Ae. aegypti saliva attenuated liver damage, as demonstrated by reduced hepatic necrosis and serum levels of alanine aminotransferase in APAP-overdosed mice. The levels of hepatic CYP2E1, the major enzyme responsible for the bioactivation of APAP, were not changed in Ae. aegypti exposed animals, suggesting no effects in the generation of hepatotoxic metabolites. On the other hand, mice treated with Ae. aegypti saliva following APAP overdose presented lower serum concentration of TNF-α, IL-6, IL-1β and IL-10, as well as reduced frequency of inflammatory cell populations in the liver, such as NKT cells, macrophages and dendritic cells. These findings show that Ae. aegypti saliva has bioactive molecules with therapeutic properties and may represent a prospective source of new compounds in the management of DILI-associated inflammatory disorders and, perhaps, many other inflammatory/autoimmune diseases.
Collapse
Affiliation(s)
- Josiane B. Assis
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruno Cogliati
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Margareth L. Capurro
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise M. Fonseca
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
18
|
Onyango MG, Ciota AT, Kramer LD. The Vector - Host - Pathogen Interface: The Next Frontier in the Battle Against Mosquito-Borne Viral Diseases? Front Cell Infect Microbiol 2020; 10:564518. [PMID: 33178624 PMCID: PMC7596266 DOI: 10.3389/fcimb.2020.564518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
An unprecedented spread of mosquito-borne viruses and increasing populations of mosquito vectors has led to an increase in the frequency of mosquito-borne virus disease outbreaks. Recent outbreaks of Zika virus (ZIKV) and yellow fever virus (YFV), among others have led to a concerted effort to understand the biology of mosquito-borne viruses and their interaction with their vector mosquito and vertebrate hosts. Recent studies have aimed to understand the vector-host-pathogen interface and how it influences infection, tropism and disease severity in the vertebrate host. The initial replication of the pathogen at the skin bite site is crucial in determining the progression of the infection in the vertebrate host. Delineating the role of the commensal microbes in the mosquito saliva as well as how they interact with the vertebrate host keratinocytes will improve our understanding of disease immunopathology and may lead to new therapeutics.
Collapse
Affiliation(s)
- Maria Gorreti Onyango
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States
| | - Alexander T Ciota
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| | - Laura D Kramer
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| |
Collapse
|
19
|
Li HH, Cai Y, Li JC, Su MP, Liu WL, Cheng L, Chou SJ, Yu GY, Wang HD, Chen CH. C-Type Lectins Link Immunological and Reproductive Processes in Aedes aegypti. iScience 2020; 23:101486. [PMID: 32891883 PMCID: PMC7481239 DOI: 10.1016/j.isci.2020.101486] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/14/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
Physiological trade-offs between mosquito immune response and reproductive capability can arise due to insufficient resource availability. C-type lectin family members may be involved in these processes. We established a GCTL-3-/- mutant Aedes aegypti using CRISPR/Cas9 to investigate the role of GCTL-3 in balancing the costs associated with immune responses to arboviral infection and reproduction. GCTL-3-/- mutants showed significantly reduced DENV-2 infection rate and gut commensal microbiota populations, as well as upregulated JAK/STAT, IMD, Toll, and AMPs immunological pathways. Mutants also had significantly shorter lifespans than controls and laid fewer eggs due to defective germ line development. dsRNA knock-down of Attacin and Gambicin, two targets of the AMPs pathway, partially rescued this reduction in reproductive capabilities. Upregulation of immune response following GCTL-3 knock-out therefore comes at a cost to reproductive fitness. Knock-out of other lectins may further improve our knowledge of the molecular and genetic mechanisms underlying reproduction-immunity trade-offs in mosquitoes.
Collapse
Affiliation(s)
- Hsing-Han Li
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, 117604, Singapore; Department of Biological Sciences, National University of Singapore, 117558, Singapore
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Matthew P Su
- Department of Biological Science, Nagoya University, Nagoya 464-8602, Japan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Lie Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Shu-Jen Chou
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Horng-Dar Wang
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan; National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan.
| |
Collapse
|
20
|
Sri-in C, Weng SC, Shiao SH, Tu WC. A simplified method for blood feeding, oral infection, and saliva collection of the dengue vector mosquitoes. PLoS One 2020; 15:e0233618. [PMID: 32469954 PMCID: PMC7259494 DOI: 10.1371/journal.pone.0233618] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/08/2020] [Indexed: 12/31/2022] Open
Abstract
A simple device using folded Parafilm-M as an artificial blood feeder was designed for studying two important dengue vector mosquitoes, Aedes aegypti and Aedes albopictus. The efficiency of the artificial blood feeder was investigated by comparing the numbers of engorged mosquitoes that fed on the artificial blood feeder versus mice as a live blood source. Significantly more engorged females Aedes aegypti fed on the artificial blood feeder than on mice. In addition, the artificial feeder could serve as a useful apparatus for oral infection via artificial blood meals, and for saliva collection in mosquitoes. Our method enabled us to collect saliva from multiple mosquitoes at once, providing sufficient infected saliva for determination of the virus titer by plaque assay analysis. Our artificial feeder has the advantage that it is simple, inexpensive, and efficient.
Collapse
Affiliation(s)
- Chalida Sri-in
- Department of Entomology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan
| | - Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wu-Chun Tu
- Department of Entomology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
21
|
Buezo Montero S, Gabrieli P, Severini F, Picci L, Di Luca M, Forneris F, Facchinelli L, Ponzi M, Lombardo F, Arcà B. Analysis in a murine model points to IgG responses against the 34k2 salivary proteins from Aedes albopictus and Aedes aegypti as novel promising candidate markers of host exposure to Aedes mosquitoes. PLoS Negl Trop Dis 2019; 13:e0007806. [PMID: 31618201 PMCID: PMC6816578 DOI: 10.1371/journal.pntd.0007806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/28/2019] [Accepted: 09/25/2019] [Indexed: 01/22/2023] Open
Abstract
Background Aedes mosquitoes are vectors of arboviral diseases of great relevance for public health. The recent outbreaks of dengue, Zika, chikungunya and the rapid worldwide spreading of Aedes albopictus emphasize the need for improvement of vector surveillance and control. Host antibody response to mosquito salivary antigens is emerging as a relevant additional tool to directly assess vector-host contact, monitor efficacy of control interventions and evaluate risk of arboviral transmission. Methodology/principal findings Groups of four BALB/c mice were immunized by exposure to bites of either Aedes albopictus or Aedes aegypti. The 34k2 salivary proteins from Ae. albopictus (al34k2) and Ae. aegypti (ae34k2) were expressed in recombinant form and Ae. albopictus salivary peptides were designed through B-cell epitopes prediction software. IgG responses to salivary gland extracts, peptides, al34k2 and ae34k2 were measured in exposed mice. Both al34k2 and ae34k2, with some individual and antigen-specific variation, elicited a clearly detectable antibody response in immunized mice. Remarkably, the two orthologous proteins showed very low level of immune cross-reactivity, suggesting they may eventually be developed as species-specific markers of host exposure. The al34k2 immunogenicity and the limited immune cross-reactivity to ae34k2 were confirmed in a single human donor hyperimmune to Ae. albopictus saliva. Conclusions/significance Our study shows that exposure to bites of Ae. albopictus or Ae. aegypti evokes in mice species-specific IgG responses to al34k2 or ae34k2, respectively. Deeper understanding of duration of antibody response and validation in natural conditions of human exposure to Aedes mosquitoes are certainly needed. However, our findings point to the al34k2 salivary protein as a promising potential candidate for the development of immunoassays to evaluate human exposure to Ae. albopictus. This would be a step forward in the establishment of a serological toolbox for the simultaneous assessment of human exposure to Aedes vectors and the pathogens they transmit. Taking advantage of several factors, as worldwide trading, climatic changes and urbanization, Aedes mosquitoes are impressively expanding their geographic distribution. A paradigm is provided by the rapid global spreading of Aedes albopictus, a species that is a competent vector of several arboviral diseases (e.g. dengue, Zika, chikungunya) and has been responsible of quite a few outbreaks in the last decade. Historically, vector control always played a pivotal role for the containment of arthropod-borne diseases, and this appears especially crucial for arboviral diseases for which no effective vaccines or specific medications are available. Currently, host exposure to mosquitoes is indirectly evaluated by entomological methods; however, exploitation of human immune responses to mosquito salivary proteins is emerging as a relevant additional tool, with important epidemiological implications for the evaluation of mosquito-borne disease risk. This study provides preliminary but solid indications that the 34k2 salivary proteins from Ae. albopictus and Aedes aegypti may be suitable candidates for the development of serological assays to evaluate spatial and/or temporal variation of human exposure to Aedes vectors. Combined to the presently available tools to assess arboviral exposure/infection, this may be of great help for the development of a serological toolbox allowing for the simultaneous determination of human exposure to Aedes vectors and to the pathogens they transmit.
Collapse
Affiliation(s)
- Sara Buezo Montero
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Paolo Gabrieli
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Italy
| | - Francesco Severini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Leonardo Picci
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Di Luca
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Federico Forneris
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Italy
| | - Luca Facchinelli
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Marta Ponzi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- * E-mail:
| |
Collapse
|