1
|
Wang Z, Nie K, Liang Y, Niu J, Yu X, Zhang O, Liu L, Shi X, Wang Y, Feng X, Zhu Y, Wang P, Cheng G. A mosquito salivary protein-driven influx of myeloid cells facilitates flavivirus transmission. EMBO J 2024; 43:1690-1721. [PMID: 38378891 PMCID: PMC11066113 DOI: 10.1038/s44318-024-00056-x] [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: 07/11/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
Mosquitoes transmit many disease-relevant flaviviruses. Efficient viral transmission to mammalian hosts requires mosquito salivary factors. However, the specific salivary components facilitating viral transmission and their mechanisms of action remain largely unknown. Here, we show that a female mosquito salivary gland-specific protein, here named A. aegypti Neutrophil Recruitment Protein (AaNRP), facilitates the transmission of Zika and dengue viruses. AaNRP promotes a rapid influx of neutrophils, followed by virus-susceptible myeloid cells toward mosquito bite sites, which facilitates establishment of local infection and systemic dissemination. Mechanistically, AaNRP engages TLR1 and TLR4 of skin-resident macrophages and activates MyD88-dependent NF-κB signaling to induce the expression of neutrophil chemoattractants. Inhibition of MyD88-NF-κB signaling with the dietary phytochemical resveratrol reduces AaNRP-mediated enhancement of flavivirus transmission by mosquitoes. These findings exemplify how salivary components can aid viral transmission, and suggest a potential prophylactic target.
Collapse
Affiliation(s)
- Zhaoyang Wang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Kaixiao Nie
- Department of Pathogen Biology, School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yan Liang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jichen Niu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Xi Yu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Oujia Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100086, China
| | - Long Liu
- Institute of Virology, Hubei University of Medicine, Shiyan, 442000, China
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Xiaolu Shi
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Yibaina Wang
- China National Center for Food Safety Risk Assessment, Beijing, 100022, China
| | - Xuechun Feng
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
| | - Yibin Zhu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China.
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
- Southwest United Graduate School, Kunming, 650092, China.
| |
Collapse
|
2
|
Pei L, Hickman HD. T Cell Surveillance during Cutaneous Viral Infections. Viruses 2024; 16:679. [PMID: 38793562 PMCID: PMC11126121 DOI: 10.3390/v16050679] [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: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
The skin is a complex tissue that provides a strong physical barrier against invading pathogens. Despite this, many viruses can access the skin and successfully replicate in either the epidermal keratinocytes or dermal immune cells. In this review, we provide an overview of the antiviral T cell biology responding to cutaneous viral infections and how these responses differ depending on the cellular targets of infection. Much of our mechanistic understanding of T cell surveillance of cutaneous infection has been gained from murine models of poxvirus and herpesvirus infection. However, we also discuss other viral infections, including flaviviruses and papillomaviruses, in which the cutaneous T cell response has been less extensively studied. In addition to the mechanisms of successful T cell control of cutaneous viral infection, we highlight knowledge gaps and future directions with possible impact on human health.
Collapse
Affiliation(s)
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
3
|
Cecilia H, Althouse BM, Azar SR, Moehn BA, Yun R, Rossi SL, Vasilakis N, Hanley KA. Aedes albopictus is not an arbovirus aficionado - Impacts of sylvatic flavivirus infection in vectors and hosts on mosquito engorgement on non-human primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.19.580944. [PMID: 38559148 PMCID: PMC10979881 DOI: 10.1101/2024.02.19.580944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The contact structure between vertebrate hosts and arthropod vectors plays a key role in the spread of arthropod-borne viruses (arboviruses); thus, it is important to determine whether arbovirus infection of either host or vector alters vector feeding behavior. Here we leveraged a study of the replication dynamics of two arboviruses isolated from their ancestral cycles in paleotropical forests, sylvatic dengue-2 (DENV-2) and Zika (ZIKV), in one non-human primate (NHP) species from the paleotropics (cynomolgus macaques, Macaca fascicularis) and one from the neotropics (squirrel monkeys, Saimiri boliviensis) to test the effect of both vector and host infection with each virus on completion of blood feeding (engorgement) of the mosquito Aedes albopictus. Although mosquitoes were starved and given no choice of hosts, engorgement rates varied dramatically, from 0% to 100%. While neither vector nor host infection systematically affected engorgement, NHP species and body temperature at the time of feeding did. We also interrogated the effect of repeated mosquito bites on cytokine expression and found that epidermal growth factor (EGF) and macrophage migration inhibitory factor (MIF) concentrations were dynamically associated with exposure to mosquito bites. This study highlights the importance of incorporating individual-level heterogeneity of vector biting in arbovirus transmission models.
Collapse
Affiliation(s)
- Hélène Cecilia
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003 USA
| | - Benjamin M. Althouse
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003 USA
- Information School, University of Washington, Seattle, WA, 98105
| | - Sasha R. Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Center for Tissue Engineering, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX 77030 USA
| | - Brett A. Moehn
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003 USA
| | - Ruimei Yun
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555 USA
| | - Shannan L. Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Department of Microbiology and Immunology, Unviersity of Texas Medical Branch, Galveston, TX 77555 USA
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, 77555 USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, 77555 USA
| | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003 USA
| |
Collapse
|
4
|
Visser I, Koenraadt CJ, Koopmans MP, Rockx B. The significance of mosquito saliva in arbovirus transmission and pathogenesis in the vertebrate host. One Health 2023. [DOI: 10.1016/j.onehlt.2023.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
|
5
|
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: 13.0] [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
|
6
|
Lu S, Danchenko M, Macaluso KR, Ribeiro JMC. Revisiting the sialome of the cat flea Ctenocephalides felis. PLoS One 2023; 18:e0279070. [PMID: 36649293 PMCID: PMC9844850 DOI: 10.1371/journal.pone.0279070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/29/2022] [Indexed: 01/18/2023] Open
Abstract
The hematophagous behaviour emerged independently in several instances during arthropod evolution. Survey of salivary gland and saliva composition and its pharmacological activity led to the conclusion that blood-feeding arthropods evolved a distinct salivary mixture that can interfere with host defensive response, thus facilitating blood acquisition and pathogen transmission. The cat flea, Ctenocephalides felis, is the major vector of several pathogens, including Rickettsia typhi, Rickettsia felis and Bartonella spp. and therefore, represents an important insect species from the medical and veterinary perspectives. Previously, a Sanger-based sialome of adult C. felis female salivary glands was published and reported 1,840 expressing sequence tags (ESTs) which were assembled into 896 contigs. Here, we provide a deeper insight into C. felis salivary gland composition using an Illumina-based sequencing approach. In the current dataset, we report 8,892 coding sequences (CDS) classified into 27 functional classes, which were assembled from 42,754,615 reads. Moreover, we paired our RNAseq data with a mass spectrometry analysis using the translated transcripts as a reference, confirming the presence of several putative secreted protein families in the cat flea salivary gland homogenates. Both transcriptomic and proteomic approaches confirmed that FS-H-like proteins and acid phosphatases lacking their putative catalytic residues are the two most abundant salivary proteins families of C. felis and are potentially related to blood acquisition. We also report several novel sequences similar to apyrases, odorant binding proteins, antigen 5, cholinesterases, proteases, and proteases inhibitors, in addition to putative novel sequences that presented low or no sequence identity to previously deposited sequences. Together, the data represents an extended reference for the identification and characterization of the pharmacological activity present in C. felis salivary glands.
Collapse
Affiliation(s)
- Stephen Lu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Rockville, Maryland, United States of America
- * E-mail:
| | - Monika Danchenko
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, United States of America
| | - Kevin R. Macaluso
- Department of Microbiology and Immunology, University of South Alabama College of Medicine, Mobile, Alabama, United States of America
| | - José M. C. Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Rockville, Maryland, United States of America
| |
Collapse
|
7
|
Wang Y, Ling L, Zhang Z, Marin-Lopez A. Current Advances in Zika Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10111816. [PMID: 36366325 PMCID: PMC9694033 DOI: 10.3390/vaccines10111816] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV), an emerging arthropod-borne flavivirus, was first isolated in Uganda in 1947 from monkeys and first detected in humans in Nigeria in 1952; it has been associated with a dramatic burden worldwide. Since then, interventions to reduce the burden of ZIKV infection have been mainly restricted to mosquito control, which in the end proved to be insufficient by itself. Hence, the situation prompted scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elucidated. Understanding the viral disease mechanism will provide a better landscape to develop prophylactic and therapeutic strategies against ZIKV. Currently, no specific vaccines or drugs have been approved for ZIKV. However, some are undergoing clinical trials. Notably, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, viral vectors, virus-like particles (VLPs), inactivated virus, live attenuated virus, peptide and protein-based vaccines, passive immunizations by using monoclonal antibodies (MAbs), and vaccines that target vector-derived antigens. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA titers, both in vitro and in vivo. This review provides a comprehensive summary of current advancements in the development of vaccines against Zika virus.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
- Correspondence:
| | - Lin Ling
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Zilei Zhang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
8
|
Marten AD, Tift CT, Tree MO, Bakke J, Conway MJ. Chronic depletion of vertebrate lipids in Aedes aegypti cells dysregulates lipid metabolism and inhibits innate immunity without altering dengue infectivity. PLoS Negl Trop Dis 2022; 16:e0010890. [PMID: 36279305 PMCID: PMC9632908 DOI: 10.1371/journal.pntd.0010890] [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: 05/23/2022] [Revised: 11/03/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Aedes aegypti is the primary vector of dengue virus (DENV) and other arboviruses. Previous literature suggests that vertebrate and invertebrate lipids and the nutritional status of mosquitoes modify virus infection. Here, we developed a vertebrate lipid-depleted Ae. aegypti cell line to investigate if chronic depletion of vertebrate lipids normally present in a blood meal and insect cell culture medium would impact cell growth and virus infection. Chronic depletion of vertebrate lipids reduced cell size and proliferation, although cells retained equivalent total intracellular lipids per cell by reducing lipolysis and modifying gene expression related to sugar and lipid metabolism. Downregulation of innate immunity genes was also observed. We hypothesized that chronic depletion of vertebrate lipids would impact virus infection; however, the same amount of DENV was produced per cell. This study reveals how Ae. aegypti cells adapt in the absence of vertebrate lipids, and how DENV can replicate equally well in cells that contain predominately vertebrate or invertebrate lipids. Aedes aegypti is a major threat to public health. Ae. aegypti is the primary vector of dengue virus types 1–4 (DENV 1–4), zika virus (ZIKV), chikungunya virus (CHIKV), and yellow fever virus (YFV). Ae. aegypti acquires arboviruses from a vertebrate host during blood feeding. Blood feeding introduces vertebrate-specific factors into the mosquito that may be important for both mosquito and virus. This study reveals that Ae. aegypti adapts to depletion of vertebrate lipids by inhibiting lipolysis and promoting de novo synthesis of invertebrate lipids, and that DENV can replicate equally well without high concentrations of cholesterol and other vertebrate lipid species. Understanding how disease vectors adapt to nutritional changes will identify novel strategies for vector control and disease mitigation.
Collapse
Affiliation(s)
- Andrew D. Marten
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan, United States of America
| | - Clara T. Tift
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan, United States of America
| | - Maya O. Tree
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan, United States of America
| | - Jesse Bakke
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan, United States of America
| | - Michael J. Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan, United States of America
- * E-mail:
| |
Collapse
|
9
|
Vander Does A, Labib A, Yosipovitch G. Update on mosquito bite reaction: Itch and hypersensitivity, pathophysiology, prevention, and treatment. Front Immunol 2022; 13:1024559. [PMID: 36211437 PMCID: PMC9532860 DOI: 10.3389/fimmu.2022.1024559] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Mosquito bites are endured by most populations worldwide. Reactions to mosquito bites range from localized wheals and papules with associated pruritus to rare systemic reactions and anaphylaxis in certain populations. The mechanism of itch is due to introduction of mosquito saliva components into the cutaneous tissue, although the exact pathophysiology is unclear. Histamine is thought to be a key player through mosquito saliva itself or through activation of mast cells by IgE or through an IgE-independent pathway. However, other salivary proteins such as tryptase and leukotrienes may induce non-histaminergic itch. Some individuals have a genetic predisposition for mosquito bites, and people with hematologic cancers, HIV, and other conditions are susceptible to robust reactions. Prevention of mosquito bites is key with physical barriers or chemical repellents. Treatment consists of second-generation antihistamines and topical corticosteroids. Further research on topical treatments that target neural-mediated itch is needed.
Collapse
|
10
|
Lu S, Martin-Martin I, Ribeiro JM, Calvo E. A deeper insight into the sialome of male and female Ochlerotatus triseriatus mosquitoes. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 147:103800. [PMID: 35787945 PMCID: PMC9494274 DOI: 10.1016/j.ibmb.2022.103800] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Over the last 20 years, advancements in sequencing technologies have highlighted the unique composition of the salivary glands of blood-feeding arthropods. Further biochemical and structural data demonstrated that salivary proteins can disrupt host hemostasis, inflammation and immunity, which favors pathogen transmission. Previously, a Sanger-based sialome of adult Ochlerotatus triseriatus female salivary glands was published based on 731 expressed sequence tag (ESTs). Here, we revisited O. triseriatus salivary gland contents using an Illumina-based sequencing approach of both male and female tissues. In the current data set, we report 10,317 DNA coding sequences classified into several functional classes. The translated transcripts also served as a reference database for proteomic analysis of O. triseriatus female saliva, in which unique peptides from 101 proteins were found. Finally, comparison of male and female libraries allowed for the identification of female-enriched transcripts that are potentially related to blood acquisition and virus transmission.
Collapse
Affiliation(s)
- Stephen Lu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jose M Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
| |
Collapse
|
11
|
Gomes R, Kolářová I, Sá-Nunes A, Carneiro M. Editorial: Hematophagous arthropod saliva: a multifunctional tool. Front Cell Infect Microbiol 2022; 12:977511. [PMID: 35909959 PMCID: PMC9326350 DOI: 10.3389/fcimb.2022.977511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Regis Gomes
- Biotechnology, Oswaldo Cruz Foundation, Eusébio, Brazil
- *Correspondence: Regis Gomes,
| | - Iva Kolářová
- Laboratory for Biology of Insect Vectors, Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Anderson Sá-Nunes
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- National Institute of Science and Technology in Molecular Entomology, National Council of Scientific and Technological Development, Rio de Janeiro, Brazil
| | - Matheus Carneiro
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
12
|
Južnič-Zonta Ž, Sanpera-Calbet I, Eritja R, Palmer JR, Escobar A, Garriga J, Oltra A, Richter-Boix A, Schaffner F, della Torre A, Miranda MÁ, Koopmans M, Barzon L, Bartumeus Ferre F. Mosquito alert: leveraging citizen science to create a GBIF mosquito occurrence dataset. GIGABYTE 2022; 2022:gigabyte54. [PMID: 36824520 PMCID: PMC9930537 DOI: 10.46471/gigabyte.54] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/12/2022] [Indexed: 11/09/2022] Open
Abstract
The Mosquito Alert dataset includes occurrence records of adult mosquitoes collected worldwide in 2014-2020 through Mosquito Alert, a citizen science system for investigating and managing disease-carrying mosquitoes. Records are linked to citizen science-submitted photographs and validated by entomologists to determine the presence of five targeted European mosquito vectors: Aedes albopictus, Ae. aegypti, Ae. japonicus, Ae. koreicus, and Culex pipiens. Most records are from Spain, reflecting Spanish national and regional funding, but since autumn 2020 substantial records from other European countries are included, thanks to volunteer entomologists coordinated by the AIM-COST Action, and to technological developments to increase scalability. Among other applications, the Mosquito Alert dataset will help develop citizen science-based early warning systems for mosquito-borne disease risk. It can also be reused for modelling vector exposure risk, or to train machine-learning detection and classification routines on the linked images, to assist with data validation and establishing automated alert systems.
Collapse
Affiliation(s)
- Živko Južnič-Zonta
- Centre d’Estudis Avançats de Blanes (CEAB-CSIC), C/d’accés a la Cala St. Francesc 14, 17300 Blanes, Girona, Spain
| | - Isis Sanpera-Calbet
- Departament de Ciències Polítiques i Socials, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Roger Eritja
- Centre de Recerca Ecològica i Aplicacions Forestals (CREAF), Edifici C Campus de, 08193 Bellaterra, Barcelona, Spain
| | - John R.B. Palmer
- Departament de Ciències Polítiques i Socials, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Agustí Escobar
- Centre de Recerca Ecològica i Aplicacions Forestals (CREAF), Edifici C Campus de, 08193 Bellaterra, Barcelona, Spain
| | - Joan Garriga
- Centre d’Estudis Avançats de Blanes (CEAB-CSIC), C/d’accés a la Cala St. Francesc 14, 17300 Blanes, Girona, Spain
| | - Aitana Oltra
- Departament de Ciències Polítiques i Socials, Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Alex Richter-Boix
- Centre de Recerca Ecològica i Aplicacions Forestals (CREAF), Edifici C Campus de, 08193 Bellaterra, Barcelona, Spain
| | - Francis Schaffner
- Francis Schaffner Consultancy (FSC), Lörracherstrasse 50, 4125 Riehen, Switzerland
| | - Alessandra della Torre
- Department Public Health and Infectious Diseases (UNIROMA1), Sapienza University, 00185 Rome, Italy
| | - Miguel Ángel Miranda
- University Balearic Islands, Applied Zoology and Animal Conservation Research Group (UIB), Ctra. Valldemossa km 7.5, 07122, Palma, Spain
| | - Marion Koopmans
- Erasmus University Medical Center (Erasmus MC), Doctor Molewaterplein 40, 3015 GD Rotterdam, Netherlands
| | - Luisa Barzon
- Department of Molecular Medicine (UNIPV), Università degli Studi di Padova, 63 Via Gabelli, 35121 Padova, Italy
| | - Frederic Bartumeus Ferre
- Centre d’Estudis Avançats de Blanes (CEAB-CSIC), C/d’accés a la Cala St. Francesc 14, 17300 Blanes, Girona, Spain,Centre de Recerca Ecològica i Aplicacions Forestals (CREAF), Edifici C Campus de, 08193 Bellaterra, Barcelona, Spain,Institució Catalana de Recerca i Estudis Avançats (ICREA), 23 Passeig de Lluís Companys, 08010 Barcelona, Spain, Corresponding author. E-mail:
| | - Mosquito Alert Digital Entomology Network
https://orcid.org/0000-0001-5319-4257Alarcón-ElbalPedro María32https://orcid.org/0000-0002-5754-862XAlexander GonzálezMikel15https://orcid.org/0000-0003-0997-3055Angeles PuigMaria31https://orcid.org/0000-0001-8818-2483Bakran-LeblKarin523https://orcid.org/0000-0002-3973-068XBalatsosGeorgios27https://orcid.org/0000-0002-8345-3229BarcelóCarlos16https://orcid.org/0000-0002-6399-4765Bengoa PaulisMikel3https://orcid.org/0000-0002-6697-302XBisiaMarina27Blanco-SierraLaura1https://orcid.org/0000-0003-3481-7310Bravo-BarrigaDaniel20https://orcid.org/0000-0002-5650-8773CaputoBeniamino14https://orcid.org/0000-0002-8085-6399CollantesFrancisco25https://orcid.org/0000-0001-6704-740XCosta OsórioHugo12Curman PosavecMarcela2https://orcid.org/0000-0002-6582-7020CvetkovikjAleksandar29https://orcid.org/0000-0001-7268-8965DeblauweIsra30https://orcid.org/0000-0001-7046-2997DelacourSarah10Escartin PeñaSanti4https://orcid.org/0000-0001-7481-4355FerragutiMartina18https://orcid.org/0000-0001-8267-6503FlacioEleonora19https://orcid.org/000-0002-4178-0133FuehrerHans-Peter23https://orcid.org/0000-0001-5236-9537GewehrSandra9https://orcid.org/0000-0002-2583-6264GunayFiliz35https://orcid.org/0000-0003-0107-5357Gutiérrez-LópezRafael16https://orcid.org/0000-0002-9582-6635HorváthCintia17https://orcid.org/0000-0002-0768-2011Ibanez-JusticiaAdolfo8https://orcid.org/0000-0002-1819-5278KadriajPerparim24https://orcid.org/0000-0001-8969-7382KalanKatja34https://orcid.org/0000-0001-5210-9727KavranMihaela21https://orcid.org/0000-0001-9775-3065KemenesiGábor22https://orcid.org/0000-0003-3464-6830KlobucarAna2https://orcid.org/0000-0001-6190-1265KuruczKornélia22https://orcid.org/0000-0001-5719-5994LongoEleonora14https://orcid.org/0000-0002-6748-9547MagallanesSergio36https://orcid.org/0000-0003-0903-8657MarianiSimone31https://orcid.org/0000-0003-2892-8583MartinouAngeliki F.6https://orcid.org/0000-0001-9945-6283Melero-AlcíbarRosario37https://orcid.org/0000-0002-3075-5020MichaelakisAntonios27https://orcid.org/0000-0002-8886-3315MicheluttiAlice11https://orcid.org/0000-0002-6003-0434MikovOgnyan28MontalvoTomas1https://orcid.org/0000-0002-5004-5763MontarsiFabrizio11PaoliFrancesca39Parrondo MontónDiego19https://orcid.org/0000-0003-1757-1822RogoziElton24https://orcid.org/0000-0001-8198-8118Ruiz-ArrondoIgnacio7https://orcid.org/0000-0002-0179-5277SeveriniFrancesco38https://orcid.org/0000-0002-7912-5791SokolovskaNikolina13https://orcid.org/0000-0003-2947-1423Sophia UnterköflerMaria23StrooArjan8https://orcid.org/0000-0003-2624-230XTeekemaSteffanie8ValsecchiAndrea1https://orcid.org/0000-0003-2463-5660VauxAlexander G. C.33https://orcid.org/0000-0001-7283-2541VeloEnkelejda24https://orcid.org/0000-0002-8963-6421ZittraCarina26Agencia de Salud Pública de Barcelona (ASPB), Plaça Lesseps 8 entresol, 08023, Barcelona, SpainAndrija Stampar Teaching Institute of Public Health (ASTIPH), Mirogojska c. 16, 10 000, Zagreb, CroatiaAnticimex Spain (Anticimex), C/ Jesús Serra Santamans, 5, Planta 3, 08174, Sant Cugat del Vallès, Barcelona, SpainAssociació Mediambiental Xatrac (Xatrac), C/ Pius Font i Quer, S/N, 17310, Lloret de Mar, Girona, SpainAustrian Agency for Health and Food Safety, Division for Public Health (AGES), Währinger Strasse 25a, 1090, Vienna, AustriaBritish Forces Cyprus, Joint Services Health Unit (JSHU), CyprusCenter for Rickettsiosis and Arthropod-Borne Diseases, Hospital Universitario San Pedro-CIBIR (CRETAV-CIBIR), C/Piqueras 98, 3° planta, 26006, La Rioja, SpainCentre for Monitoring of Vectors, National Reference Centre, Netherlands Food and Consumer Product Safety Authority (CMV-NVWA), Geertjesweg 15, 6706 EA, Wageningen, NetherlandsEcodevelopment S.A. (ECODEV), Thesi Mezaria, PO Box 2420, 57010 Filyro, GreeceUniversity of Zaragoza, Faculty of Veterinary Medicine of Zaragoza, Animal Health Department (UNIZAR), C/ Miguel Servet 177, 50013, Zaragoza, SpainIstituto Zooprofilattico Sperimentale delle Venezie (IZSVe), Viale dell’Università 10, 35020, Legnaro (Padua), ItalyNational Institute of Health, Centre for Vectors and Infectious Diseases Research (INSA-CEVDI), Avenida Padre Cruz, 1649-016, Lisboa, PortugalPHI Center for Public Health-Skopje (CPH), blv.3rd Macedonian brigade, no.18, Skopje, North MacedoniaSapienza University, Department Public Health and Infectious Diseases (UNIROMA1), Piazzale Aldo Moro 5, 00198, Rome, ItalyUniversidad Iberoamericana (UNIBE), Avenida Francia 129, 10203, Santo Domingo, Dominican RepublicUniversity Balearic Islands, Applied Zoology and Animal Conservation Research Group (UIB), Ctra. Valldemossa km 7.5, 07122, Palma, SpainUniversity of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca (USAMV-CN), Calea Mănăştur 3-5, Cluj-Napoca, 400372, RomaniaUniversity of Amsterdam, Department of Theoretical and Computational Ecology, Institute for Biodiversity and Ecosystem Dynamics (UvA), Science Park 904, 1098XH, Amsterdam, NetherlandsUniversity of Applied Scieces and Arts of Southern Switzerland, Institute of Microbiology (SUPSI), Via Flora Ruchat-Roncati 15, 6850, Mendrisio Switzerland, SwitzerlandUniversity of Extremadura, Veterinary Faculty, Department of Animal Health (Uex), Av/ Universidad S/N 10003 Cáceres,
SpainUniversity of Novi Sad, Faculty of Agriculture, Laboratory for Medical and Veterinary Entomology (UNSFA), Trg Dositeja Obradovića 8, 21000, Novi Sad, SerbiaUniversity of Pécs (UP), Ifúság útja 6, 7624, Pécs, HungaryUniversity of Veterinary Medicine Vienna, Institute of Parasitology (Vetmeduni), Veterinärplatz 1, 1210, Vienna, AustriaInstitute of Public Health, Department of Epidemiology and Control of Infectious Diseases, Vectors’ Control Unit (IPH), Str: “Aleksander Moisiu”, No. 80, Tirana, AlbaniaUniversidad de Murcia, Departamento de Zoología y Antropología Física (UM), Campus de Espinardo, 30100 Murcia, SpainUniversity of Vienna, Department of Functional and Evolutionary Ecology (UNIVIE), Djerassiplatz 1, 1030, Vienna, AustriaBenaki Phytopathological Institute, Laboratory of Insects and Parasites of Medical Importance (BPI), 8, Stefanou Delta str., 14561 Kifissia, Athens, GreeceNational Centre of Infectious and Parasitic Diseases (NCIPD), 26, Yanko Sakazov blvd., 1504, Sofia, BulgariaSs. Cyril and Methodius University in Skopje, Faculty of Veterinary Medicine-Skopje (FVMS), Lazar Pop-Trajkov 5-7, 1000, Skopje, North MacedoniaInstitute of Tropical Medicine, Department of Biomedical Sciences, Unit of Entomology (ITM), Nationalestraat 155, 2000, Antwerp, BelgiumCentre d’Estudis Avançats de Blanes (CEAB-CSIC), C/ d’accés a la Cala St. Francesc 14, 17300 Blanes, Girona, SpainUniversidad Cardenal Herrera CEU-CEU Universities, Facultad de Veterinaria, Veterinary Public Health and Food Science and Technology, Department of Animal Production and Health (PASAPTA), C/ Tirant lo Blanc, 7, 46115 Alfara del Patriarca, Valencia, SpainMedical Entomology, UK Health Security Agency (UKHSA), Porton Down, Salisbury, SP4 0JG, United KingdomUniversity of Primorska, Faculty of Mathematics, Natural Sciences and Information Technologies (UP FAMNIT), Glagoljaška ulica 8, 6000, Koper, SloveniaHacettepe University, Department of Biology, Ecology Section, Vector Ecology Research Group (HU-VERG), Hacettepe University, Beytepe Campus, 06800, Ankara, TurkeyEstación Biológica de Doñana, Departamento de Ecología de los Humedales (EBD-CSIC), Avda. Américo Vespucio 26, 41092, Sevilla, SpainCentro de Educación Superior Hygiea (HYGIEA), Av. de Pablo VI, 9, 28223, Pozuelo de Alarcón, Madrid, SpainIstituto Superiore di Sanità, Department of Infectious Diseases (ISS), Viale Regina Elena, 299, 00161, Roma, ItalyMuseo di Scienze di Trento (MUSE), Corso del Lavoro e della Scienza, 3, 38122, Trento, Italy
| | | |
Collapse
|
13
|
Vector-Borne Viral Diseases as a Current Threat for Human and Animal Health—One Health Perspective. J Clin Med 2022; 11:jcm11113026. [PMID: 35683413 PMCID: PMC9181581 DOI: 10.3390/jcm11113026] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Over the last decades, an increase in the emergence or re-emergence of arthropod-borne viruses has been observed in many regions. Viruses such as dengue, yellow fever, or zika are a threat for millions of people on different continents. On the other hand, some arboviruses are still described as endemic, however, they could become more important in the near future. Additionally, there is a group of arboviruses that, although important for animal breeding, are not a direct threat for human health. Those include, e.g., Schmallenberg, bluetongue, or African swine fever viruses. This review focuses on arboviruses and their major vectors: mosquitoes, ticks, biting midges, and sandflies. We discuss the current knowledge on arbovirus transmission, ecology, and methods of prevention. As arboviruses are a challenge to both human and animal health, successful prevention and control are therefore only possible through a One Health perspective.
Collapse
|
14
|
Shinzawa N, Kashima C, Aonuma H, Takahashi K, Shimojima M, Fukumoto S, Saiki E, Yamamoto DS, Yoshida S, Matsuoka H, Kawaoka Y, Kanuka H. Generation of Transgenic Mosquitoes Harboring a Replication-Restricted Virus. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.850111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Live microbe vaccines are designed to elicit strong cellular and antibody responses without developing the symptoms of the disease, and these are effective in preventing infectious diseases. A flying vaccinator (also known as a flying syringe) is a conceptual, genetically engineered hematophagous insect that is used to deliver vaccines such as an antigen from a parasite produced in mosquito saliva; bites from such insects may elicit antibody production by immunizing the host with an antigen through blood-feeding. In addition to a simple vaccine antigen, a flying vaccinator may potentially load a live attenuated microbe with an appropriate mechanism for sustaining its constitutive proliferation in the insect. In this study, a recombinant vesicular stomatitis virus (VSV) lacking the glycoprotein gene (VSV-G) was used to produce replication-restricted VSV (rrVSV) containing GFP. Transgenic Anopheles stephensi mosquitoes, in which the salivary glands expressed a VSV-G gene driven by an aapp salivary gland-specific promoter, were generated and injected intraperitoneally with rrVSV. The injected rrVSV entered the cells of the salivary gland and stimulated endogenous production of progeny rrVSV particles, as seen in rrVSV-infected Drosophila melanogaster expressing VSV-G. These data suggested the possibility of developing a valuable tool for delivering genetically attenuated virus vaccines via mosquito saliva, although efficient replication-restricted virus production is required.
Collapse
|
15
|
Aleshnick M, Florez-Cuadros M, Martinson T, Wilder BK. Monoclonal antibodies for malaria prevention. Mol Ther 2022; 30:1810-1821. [PMID: 35395399 PMCID: PMC8979832 DOI: 10.1016/j.ymthe.2022.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/04/2022] [Accepted: 04/01/2022] [Indexed: 11/29/2022] Open
Abstract
Monoclonal antibodies are highly specific proteins that are cloned from a single B cell and bind to a single epitope on a pathogen. These laboratory-made molecules can serve as prophylactics or therapeutics for infectious diseases and have an impressive capacity to modulate the progression of disease, as demonstrated for the first time on a large scale during the COVID-19 pandemic. The high specificity and natural starting point of monoclonal antibodies afford an encouraging safety profile, yet the high cost of production remains a major limitation to their widespread use. While a monoclonal antibody approach to abrogating malaria infection is not yet available, the unique life cycle of the malaria parasite affords many opportunities for such proteins to act, and preliminary research into the efficacy of monoclonal antibodies in preventing malaria infection, disease, and transmission is encouraging. This review examines the current status and future outlook for monoclonal antibodies against malaria in the context of the complex life cycle and varied antigenic targets expressed in the human and mosquito hosts, and provides insight into the strengths and limitations of this approach to curtailing one of humanity’s oldest and deadliest diseases.
Collapse
Affiliation(s)
- Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Thomas Martinson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Brandon K Wilder
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA; Department of Parasitology, U.S. Naval Medical Research 6 (NAMRU-6), Lima, Peru
| |
Collapse
|
16
|
Li Z, Soohoo-Hui A, O’Hara FM, Swale DR. ATP-sensitive inward rectifier potassium channels reveal functional linkage between salivary gland function and blood feeding in the mosquito, Aedes aegypti. Commun Biol 2022; 5:278. [PMID: 35347209 PMCID: PMC8960802 DOI: 10.1038/s42003-022-03222-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/02/2022] [Indexed: 12/03/2022] Open
Abstract
Reducing saliva secretions into the vertebrate host reduces feeding efficacy by most hematophagous arthropods. However, seminal studies suggested saliva is not a prerequisite for blood feeding in Aedes aegypti. To test this paradigm, we manually transected the salivary duct of female A. aegypti and an inability to salivate was correlated to an inability to imbibe blood. These data justified testing the relevance of inwardly rectifying potassium (Kir) channels in the A. aegypti salivary gland as an antifeedant target site. Pharmacological activation of ATP-gated Kir (KATP) channels reduced the secretory activity of the salivary gland by 15-fold that led to near elimination of blood ingestion during feeding. The reduced salivation and feeding success nearly eliminated horizontal transmission and acquisition of Dengue virus-2 (DENV2). These data suggest mosquito salivation is a prerequisite for blood feeding and provide evidence that KATP channels are critical for salivation, feeding, and vector competency. The salivary gland of Aedes aegypti is needed for efficient blood feeding, and disruption of ATP-gated Kir channels prevents salivation and blood feeding in A. aegypti as well as horizontal transmission and acquisition of Dengue virus2.
Collapse
|
17
|
A leafhopper saliva protein mediates horizontal transmission of viral pathogens from insect vectors into rice phloem. Commun Biol 2022; 5:204. [PMID: 35246603 PMCID: PMC8897447 DOI: 10.1038/s42003-022-03160-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022] Open
Abstract
Numerous insects transmit viruses together with saliva to plant phloem, but the roles of saliva components remain elusive. Here, we report that calcium-binding protein (CBP), a universal insect saliva protein, is modified to benefit horizontal transmission of a devastating rice reovirus into plant phloem. CBP effectively competes with virus-induced filaments to target and traverse actin-based apical plasmalemma into saliva-stored cavities in salivary glands of leafhopper vector. Thus, the inhibition of CBP expression by viral infection facilitates filament-mediated viral secretion into salivary cavities and then into plant phloem. Furthermore, virus-mediated reduction of CBP secretion causes an increase of cytosolic Ca2+ levels in rice, triggering substantial callose deposition and H2O2 production. Thus, viruliferous vectors encounter stronger feeding barriers, probe more frequently, and secrete more saliva into plants, ultimately enhancing viral transmission. We thus conclude that the inhibition of CBP secretion facilitates viral secretion and increases host defense response to benefit viral transmission. CBP, a calcium binding protein found in insect saliva, allows for the transmission of the devastating rice gall dwarf virus into plant phloem. This interaction with CBP is compounded by stronger feeding barriers, more frequent probing behavior, and increased saliva secretion into plants by insect vectors, all increasing the likelihood of viral transmission.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review provides an overview of arthropod-borne virus (arbovirus) infections that are important causes of human neurological infections world-wide. As many of the individual viruses in a specific genus or family cause overlapping clinical syndromes, this review discusses important viruses in groups to highlight some of the similarities and differences in groups of neuroinvasive arbovirus infections. RECENT FINDINGS Arboviruses that cause neurological infections in humans continue to emerge and distribute to new regions. The geographic range of the vectors, the hosts and subsequent arbovirus infections in humans continues to expand and evolve. As emerging arboviruses move into new geographic regions, it is important to examine the associated epidemiological and clinical impacts of these infections as they enter new populations. SUMMARY Arboviruses from the Flaviviridae, Togaviridae and Bunyaviridae families continue to emerge and spread into new regions. The arboviruses within these virus families cause characteristic neuroinvasive diseases in human populations. A complete understanding of the epidemiological and clinical features of the neuroinvasive arboviruses is important such that these pathogens can be recognized and diagnosed in humans as they emerge. Ongoing research to develop rapid, accurate diagnostics, therapeutic options and vaccines for these pathogens is needed to address future outbreaks of disease in human populations.
Collapse
|
19
|
Cimica V, Galarza JM, Rashid S, Stedman TT. Current development of Zika virus vaccines with special emphasis on virus-like particle technology. Expert Rev Vaccines 2021; 20:1483-1498. [PMID: 34148481 DOI: 10.1080/14760584.2021.1945447] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Zika virus disease received little attention until its recent explosive emergence around the globe. The devastating consequences of this pandemic include congenital Zika syndrome (CZS) and the neurological autoimmune disorder Guillain-Barré syndrome. These potential outcomes prompted massive efforts to understand the course of Zika infection and to develop therapeutic and prophylactic strategies for treatment and prevention of disease.Area covered: Preclinical and clinical data demonstrate that a safe and efficacious vaccine for protection against Zika virus infection is possible in the near future. Nevertheless, significant knowledge gaps regarding the outcome of a mass vaccination strategy exist and must be addressed. Zika virus circulates in flavivirus-endemic regions, an ideal Zika vaccine should avoid the potential of antibody-dependent enhancement from exposure to dengue virus. Prevention of CZS is the primary goal for immunization, and the vaccine must provide protection against intrauterine transmission for use during pregnancy and in women of childbearing age. Ideally, a vaccine should also prevent sexual transmission of the virus through mucosal protection.Expert opinion: This review describes current vaccine approaches against Zika virus with particular attention to the application of virus-like particle (VLP) technology as a strategy for solving the challenges of Zika virus immunization.
Collapse
Affiliation(s)
- Velasco Cimica
- American Type Culture Collection (ATCC), Manassas, VA, USA
| | | | - Sujatha Rashid
- American Type Culture Collection (ATCC), Manassas, VA, USA
| | | |
Collapse
|
20
|
Hibl BM, Dailey Garnes NJM, Kneubehl AR, Vogt MB, Spencer Clinton JL, Rico-Hesse RR. Mosquito-bite infection of humanized mice with chikungunya virus produces systemic disease with long-term effects. PLoS Negl Trop Dis 2021; 15:e0009427. [PMID: 34106915 PMCID: PMC8189471 DOI: 10.1371/journal.pntd.0009427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/02/2021] [Indexed: 12/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging, mosquito-borne alphavirus responsible for acute to chronic arthralgias and neuropathies. Although it originated in central Africa, recent reports of disease have come from many parts of the world, including the Americas. While limiting human CHIKV cases through mosquito control has been used, it has not been entirely successful. There are currently no licensed vaccines or treatments specific for CHIKV disease, thus more work is needed to develop effective countermeasures. Current animal research on CHIKV is often not representative of human disease. Most models use CHIKV needle inoculation via unnatural routes to create immediate viremia and localized clinical signs; these methods neglect the natural route of transmission (the mosquito vector bite) and the associated human immune response. Since mosquito saliva has been shown to have a profound effect on viral pathogenesis, we evaluated a novel model of infection that included the natural vector, Aedes species mosquitoes, transmitting CHIKV to mice containing components of the human immune system. Humanized mice infected by 3-6 mosquito bites showed signs of systemic infection, with demonstrable viremia (by qRT-PCR and immunofluorescent antibody assay), mild to moderate clinical signs (by observation, histology, and immunohistochemistry), and immune responses consistent with human infection (by flow cytometry and IgM ELISA). This model should give a better understanding of human CHIKV disease and allow for more realistic evaluations of mechanisms of pathogenesis, prophylaxis, and treatments.
Collapse
Affiliation(s)
- Brianne M. Hibl
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Natalie J. M. Dailey Garnes
- Section of Infectious Disease, Department of Internal Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander R. Kneubehl
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Megan B. Vogt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca R. Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
21
|
Demarta-Gatsi C, Mécheri S. Vector saliva controlled inflammatory response of the host may represent the Achilles heel during pathogen transmission. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200155. [PMID: 34035796 PMCID: PMC8128132 DOI: 10.1590/1678-9199-jvatitd-2020-0155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Infection with vector-borne pathogens starts with the inoculation of these pathogens during blood feeding. In endemic regions, the population is regularly bitten by naive vectors, implicating a permanent stimulation of the immune system by the vector saliva itself (pre-immune context). Comparatively, the number of bites received by exposed individuals from non-infected vectors is much higher than the bites from infected ones. Therefore, vector saliva and the immunological response in the skin may play an important role, so far underestimated, in the establishment of anti-pathogen immunity in endemic areas. Hence, the parasite biology and the disease pathogenesis in “saliva-primed” and “saliva-unprimed” individuals must be different. This integrated view on how the pathogen evolves within the host together with vector salivary components, which are known to be endowed with a variety of pharmacological and immunological properties, must remain the focus of any investigational study dealing with vector-borne diseases. Considering this three-way partnership, the host skin (immune system), the pathogen, and the vector saliva, the approach that consists in the validation of vector saliva as a source of molecular entities with anti-disease vaccine potential has been recently a subject of active and fruitful investigation. As an example, the vaccination with maxadilan, a potent vasodilator peptide extracted from the saliva of the sand fly Lutzomyia longipalpis, was able to protect against infection with various leishmanial parasites. More interestingly, a universal mosquito saliva vaccine that may potentially protect against a range of mosquito-borne infections including malaria, dengue, Zika, chikungunya and yellow fever. In this review, we highlight the key role played by the immunobiology of vector saliva in shaping the outcome of vector-borne diseases and discuss the value of studying diseases in the light of intimate cross talk among the pathogen, the vector saliva, and the host immune mechanisms.
Collapse
Affiliation(s)
- Claudia Demarta-Gatsi
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France.,Medicines for Malaria Venture (MMV), Geneva, Switzerland.,Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France
| | - Salah Mécheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France
| |
Collapse
|
22
|
Pierson BC, Cardile AP, Okwesili AC, Downs IL, Reisler RB, Boudreau EF, Kortepeter MG, Koca CD, Ranadive MV, Petitt PL, Kanesa-Thasan N, Rivard RG, Liggett DL, Haller JM, Norris SL, Purcell BK, Pittman PR, Saunders DL, Keshtkar Jahromi M. Safety and immunogenicity of an inactivated eastern equine encephalitis virus vaccine. Vaccine 2021; 39:2780-2790. [PMID: 33888325 DOI: 10.1016/j.vaccine.2021.03.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Eastern equine encephalitis virus (EEEV) is a mosquito borne alphavirus spread primarily in Atlantic and Gulf Coast regions of the United States. EEEV is the causative agent of a devastating meningoencephalitis syndrome, with approximately 30% mortality and significant morbidity. There is no licensed human vaccine against EEEV. An inactivated EEEV vaccine has been offered under investigational new drug (IND) protocols at the United States Army Medical Research Institute of Infectious Diseases (USAMRIID) since 1976. METHODS Healthy at-risk laboratory personnel received inactivated PE-6 strain EEEV (TSI-GSD 104) vaccine under two separate IND protocols. Protocol FY 99-11 (2002-2008) had a primary series consisting of doses on day 0, 7, and 28. Protocol FY 06-31 (2008-2016) utilized a primary series with doses on day 0 and 28, and month 6. Participants with an inadequate immune response, plaque reduction neutralization test with 80% cut-off (PRNT80) titer < 40, received booster vaccination. Volunteers with prior EEEV vaccination were eligible to enroll for booster doses based on annual titer evaluation. RESULTS The FY06-31 dosing schema resulted in significantly greater post-primary series immune response (PRNT80 ≥ 40) rates (84% vs 54%) and geometric mean titers (184.1 vs 39.4). The FY 06-31 dosing schema also resulted in significantly greater cumulative annual immune response rates from 1 to up to 7 years post vaccination (75% vs 59%) and geometric mean of titers (60.1 vs 43.0). The majority of probably or definitely related adverse events were mild and local; there were no probably or definitely related serious adverse events. CONCLUSIONS Inactivated PE-6 EEEV vaccine is safe and immunogenic in at-risk laboratory personnel. A prolonged primary series, with month 6 dose, significantly improved vaccine immunogenicity both post-primary series and longitudinally on annual titers. Despite decades of safe use under IND, full licensure is not planned due to manufacturing constraints, and ongoing development of alternatives.
Collapse
Affiliation(s)
- Benjamin C Pierson
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States.
| | - Anthony P Cardile
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Arthur C Okwesili
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Isaac L Downs
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Ronald B Reisler
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Ellen F Boudreau
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Mark G Kortepeter
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Craig D Koca
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Manmohan V Ranadive
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Patricia L Petitt
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Niranjan Kanesa-Thasan
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Robert G Rivard
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Dani L Liggett
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Jeannine M Haller
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Sarah L Norris
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Bret K Purcell
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Phillip R Pittman
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - David L Saunders
- Division of Medicine, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Frederick, MD 21702, United States
| | - Maryam Keshtkar Jahromi
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205, United States
| |
Collapse
|
23
|
Olajiga O, Holguin-Rocha AF, Rippee-Brooks M, Eppler M, Harris SL, Londono-Renteria B. Vertebrate Responses against Arthropod Salivary Proteins and Their Therapeutic Potential. Vaccines (Basel) 2021; 9:347. [PMID: 33916367 PMCID: PMC8066741 DOI: 10.3390/vaccines9040347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 01/11/2023] Open
Abstract
The saliva of hematophagous arthropods contains a group of active proteins to counteract host responses against injury and to facilitate the success of a bloodmeal. These salivary proteins have significant impacts on modulating pathogen transmission, immunogenicity expression, the establishment of infection, and even disease severity. Recent studies have shown that several salivary proteins are immunogenic and antibodies against them may block infection, thereby suggesting potential vaccine candidates. Here, we discuss the most relevant salivary proteins currently studied for their therapeutic potential as vaccine candidates or to control the transmission of human vector-borne pathogens and immune responses against different arthropod salivary proteins.
Collapse
Affiliation(s)
- Olayinka Olajiga
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Andrés F. Holguin-Rocha
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | | | - Megan Eppler
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Shanice L. Harris
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Berlin Londono-Renteria
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| |
Collapse
|
24
|
Perspectives on New Vaccines against Arboviruses Using Insect-Specific Viruses as Platforms. Vaccines (Basel) 2021; 9:vaccines9030263. [PMID: 33809576 PMCID: PMC7999276 DOI: 10.3390/vaccines9030263] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/07/2021] [Accepted: 03/13/2021] [Indexed: 11/17/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are global pathogens circulating endemically with local explosive outbreaks and constant encroachment into new locations. Few vaccines against arboviruses exist; most for humans are in development or clinical trials. Insect-specific viruses (ISVs) offer a unique platform for expression of arbovirus proteins, through the creation of ISV/arbovirus chimeras. Studies have shown promising results of these vaccines with several advantages over their wild-type counterparts. In this review, we discuss the current status of these potential vaccines using ISVs.
Collapse
|
25
|
Deep learning identification for citizen science surveillance of tiger mosquitoes. Sci Rep 2021; 11:4718. [PMID: 33633197 PMCID: PMC7907246 DOI: 10.1038/s41598-021-83657-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/27/2021] [Indexed: 11/18/2022] Open
Abstract
Global monitoring of disease vectors is undoubtedly becoming an urgent need as the human population rises and becomes increasingly mobile, international commercial exchanges increase, and climate change expands the habitats of many vector species. Traditional surveillance of mosquitoes, vectors of many diseases, relies on catches, which requires regular manual inspection and reporting, and dedicated personnel, making large-scale monitoring difficult and expensive. New approaches are solving the problem of scalability by relying on smartphones and the Internet to enable novel community-based and digital observatories, where people can upload pictures of mosquitoes whenever they encounter them. An example is the Mosquito Alert citizen science system, which includes a dedicated mobile phone app through which geotagged images are collected. This system provides a viable option for monitoring the spread of various mosquito species across the globe, although it is partly limited by the quality of the citizen scientists’ photos. To make the system useful for public health agencies, and to give feedback to the volunteering citizens, the submitted images are inspected and labeled by entomology experts. Although citizen-based data collection can greatly broaden disease-vector monitoring scales, manual inspection of each image is not an easily scalable option in the long run, and the system could be improved through automation. Based on Mosquito Alert’s curated database of expert-validated mosquito photos, we trained a deep learning model to find tiger mosquitoes (Aedes albopictus), a species that is responsible for spreading chikungunya, dengue, and Zika among other diseases. The highly accurate 0.96 area under the receiver operating characteristic curve score promises not only a helpful pre-selector for the expert validation process but also an automated classifier giving quick feedback to the app participants, which may help to keep them motivated. In the paper, we also explored the possibilities of using the model to improve future data collection quality as a feedback loop.
Collapse
|
26
|
Marin-Lopez A, Wang Y, Jiang J, Ledizet M, Fikrig E. AgBR1 and NeSt1 antisera protect mice from Aedes aegypti-borne Zika infection. Vaccine 2021; 39:1675-1679. [PMID: 33622591 DOI: 10.1016/j.vaccine.2021.01.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/26/2021] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
Zika virus(ZIKV) is primarily spread by Aedes. aegyptimosquitoes. Infection with ZIKV can result in diverse clinical symptoms in humans, ranging from mild to severe. Previously, we demonstrated that passive immunization against A. aegypti AgBR1 or NeSt1 antiserum, two mosquito saliva proteins that are transmitted with the virus, conferred partial protection against ZIKV in mice. Each individual antiserum altered the early host response in the skin and reduced viremia. Here, we show that passive immunization with a combination of AgBR1- and NeSt1-specific antibodies enhanced survival and reduced the viral burden in blood, thereby protecting mice from mosquito-borne ZIKV infection. This finding suggests that targeting a combination of mosquito saliva proteins, with AgBR1 and NeSt1 as model antigens, may be used as a vaccine strategy to help prevent mosquito-borne ZIKV infection.
Collapse
Affiliation(s)
- Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Yuchen Wang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, Hubei 430072, China.
| | - Junjun Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | | | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
27
|
Abstract
Efforts to produce vaccines against SARS and MERS were prematurely halted since their scope was perceived to be geographically restricted and they were subsequently categorized as neglected diseases. However, when a similar virus spread globally triggering the COVID-19 pandemic, we were harshly reminded that several other neglected diseases might also be waiting for the perfect opportunity to become mainstream. As climate change drives urbanization, natural selection of pathogens and their intermediate vectors and reservoirs, the risk of neglected diseases emerging within a larger susceptible pool becomes an even greater threat. Availability of a vaccine for COVID-19 is widely considered the only way to end this pandemic. Similarly, vaccines are also seen as the best tools available to control the spread of neglected (sometimes referred to as emerging or re-emerging) diseases, until the water, hygiene and sanitation infrastructure is improved in areas of their prevalence. Vaccine production is usually cost and labour intensive and thus minimal funding is directed towards controlling and eliminating neglected diseases (NDs). A customised but sustainable approach is needed to develop and deploy vaccines against NDs. While safety, efficacy and public trust are the three main success pillars for most vaccines, affordability is vital when formulating vaccines for neglected diseases.
Collapse
|
28
|
Shrivastava G, Valenzuela Leon PC, Calvo E. Inflammasome Fuels Dengue Severity. Front Cell Infect Microbiol 2020; 10:489. [PMID: 33014899 PMCID: PMC7511630 DOI: 10.3389/fcimb.2020.00489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
Dengue is an acute febrile disease triggered by dengue virus. Dengue is the widespread and rapidly transmitted mosquito-borne viral disease of humans. Diverse symptoms and diseases due to Dengue virus (DENV) infection ranges from dengue fever, dengue hemorrhagic fever (life-threatening) and dengue shock syndrome characterized by shock, endothelial dysfunction and vascular leakage. Several studies have linked the severity of dengue with the induction of inflammasome. DENV activates the NLRP3-specific inflammasome in DENV infected human patients, mice; specifically, mouse bone marrow derived macrophages (BMDMs), dendritic cells, endothelial cells, human peripheral blood mononuclear cells (PBMCs), keratinocytes, monocyte-differentiated macrophages (THP-1), and platelets. Dengue virus mediated inflammasome initiates the maturation of IL-1β and IL-18, which are critical for dengue pathology and inflammatory response. Several studies have reported the molecular mechanism through which (host and viral factors) dengue induces inflammasome, unravels the possible mechanisms of DENV pathogenesis and sets up the stage for the advancement of DENV therapeutics. In this perspective article, we discuss the potential implications and our understanding of inflammasome mechanisms of dengue virus and highlight research areas that have potential to inhibit the pathogenesis of viral diseases, specifically for dengue.
Collapse
Affiliation(s)
- Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
29
|
Leandro ADS, Rios JA, Britto ADS, Galvão SR, Lopes RD, Rivas AV, Martins CA, da Silva I, Delai RM, Gonçalves DD, da Silva MAN, Palacio-Cortès AM, Schuartz V, Sibim AC, de Castro WAC. Malathion insecticide resistance in Aedes aegypti: laboratory conditions and in situ experimental approach through adult entomological surveillance. Trop Med Int Health 2020; 25:1271-1282. [PMID: 32746492 DOI: 10.1111/tmi.13474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE In Brazil, the most common method of controlling outbreaks of arbovirus is by the use of chemical sprays, which kill the insect vector, Aedes aegypti. The main objective of this study was to evaluate the resistance of Ae. aegypti to the insecticide, malathion, in situ. The location of this study was the municipality of Foz do Iguaçu, in the state of Paraná, Brazil. METHODS Ultra-low-volume (ULV) fogging equipment was used, by vehicle, to apply the insecticide in situ, and mosquito populations after treatment were compared with those of control areas. The resistance of strains collected from the municipality was compared to the Rockefeller strain under laboratory conditions. RESULTS We found 220 adult female specimens and 7423 eggs of Ae. aegypti in the areas subjected to UBV treatment, whereas 245 adult females and 10 557 eggs were found in the control areas. The UBV treatment area showed no significant difference compared to the control area, for all the indices. Mortality of the Rockefeller colony varied more quickly when there were slight variations in malathion concentration than the Foz do Iguaçu population.
Collapse
Affiliation(s)
| | - Jean Avemir Rios
- Zoonoses Surveillance Unit, Municipal Secretary of Health, Foz do Iguaçu, Brazil
| | | | | | - Renata Defante Lopes
- Zoonoses Surveillance Unit, Municipal Secretary of Health, Foz do Iguaçu, Brazil
| | - Açucena Veleh Rivas
- Latin-American Institute of Life Sciences and Nature, Federal University of Latin American Integration, Foz do Iguaçu, Brazil.,One Health Laboratory at the Three-Border Tropical Medicine Center, Itaiguapy Foundation - Institute of Teaching and Research, Foz do Iguaçu, Brazil
| | | | - Isaac da Silva
- Zoonoses Surveillance Unit, Municipal Secretary of Health, Foz do Iguaçu, Brazil
| | - Robson Michael Delai
- One Health Laboratory at the Three-Border Tropical Medicine Center, Itaiguapy Foundation - Institute of Teaching and Research, Foz do Iguaçu, Brazil.,Department of Preventive Veterinary Medicine and Public Health, Paranaense University, Umuarama, Brazil
| | - Daniela Dib Gonçalves
- Department of Preventive Veterinary Medicine and Public Health, Paranaense University, Umuarama, Brazil
| | | | - Angela Maria Palacio-Cortès
- Laboratory of Morphology and Physiology of Culicidae and Chironomidae, Federal University of Paraná, Curitiba, Brazil
| | - Valéria Schuartz
- Laboratory of Morphology and Physiology of Culicidae and Chironomidae, Federal University of Paraná, Curitiba, Brazil
| | - Alessandra Cristiane Sibim
- Latin-American Institute of Technology, Infrastructure and Territory, Federal University of Latin American Integration, Foz do Iguaçu, Brazil
| | - Wagner Antonio Chiba de Castro
- Latin-American Institute of Life Sciences and Nature, Federal University of Latin American Integration, Foz do Iguaçu, Brazil
| |
Collapse
|
30
|
Londono-Renteria B, Montiel J, Calvo E, Tobón-Castaño A, Valdivia HO, Escobedo-Vargas K, Romero L, Bosantes M, Fisher ML, Conway MJ, Vásquez GM, Lenhart AE. Antibody Responses Against Anopheles darlingi Immunogenic Peptides in Plasmodium Infected Humans. Front Cell Infect Microbiol 2020; 10:455. [PMID: 32984076 PMCID: PMC7488213 DOI: 10.3389/fcimb.2020.00455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/24/2020] [Indexed: 11/15/2022] Open
Abstract
Introduction: Malaria is still an important vector-borne disease in the New World tropics. Despite the recent decline in malaria due to Plasmodium falciparum infection in Africa, a rise in Plasmodium infections has been detected in several low malaria transmission areas in Latin America. One of the main obstacles in the battle against malaria is the lack of innovative tools to assess malaria transmission risk, and the behavioral plasticity of one of the main malaria vectors in Latin America, Anopheles darlingi. Methods: We used human IgG antibodies against mosquito salivary gland proteins as a measure of disease risk. Whole salivary gland antigen (SGA) from Anopheles darlingi mosquitoes was used as antigen in Western blot experiments, in which a ~65 kDa protein was visualized as the main immunogenic band and sent for sequencing by mass spectrometry. Apyrase and peroxidase peptides were designed and used as antigens in an ELISA-based test to measure human IgG antibody responses in people with different clinical presentations of malaria. Results: Liquid chromatography–mass spectrometry revealed 17 proteins contained in the ~65 kDa band, with an apyrase and a peroxidase as the two most abundant proteins. Detection of IgG antibodies against salivary antigens by ELISA revealed a significant higher antibody levels in people with malaria infection when compared to uninfected volunteers using the AnDar_Apy1 and AnDar_Apy2 peptides. We also detected a significant positive correlation between the anti-peptides IgG levels and antibodies against the Plasmodium vivax and P. falciparum antigens PvMSP1 and PfMSP1. Odd ratios suggest that people with higher IgG antibodies against the apyrase peptides were up to five times more likely to have a malaria infection. Conclusion: Antibodies against salivary peptides from An. darlingi salivary gland proteins may be used as biomarkers for malaria risk.
Collapse
Affiliation(s)
- Berlin Londono-Renteria
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Jehidys Montiel
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergies and Infectious Diseases (NIAID/NIH), Rockville, MD, United States
| | | | - Hugo O Valdivia
- U.S. Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | | | - Luz Romero
- U.S. Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru.,Asociación Benéfica PRISMA, Lima, Peru
| | - Maria Bosantes
- U.S. Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru.,Asociación Benéfica PRISMA, Lima, Peru
| | | | - Michael J Conway
- Central Michigan University College of Medicine, Mount Pleasant, MI, United States
| | | | - Audrey E Lenhart
- Division of Parasitic Diseases and Malaria, Entomology Branch, Centers for Disease Control and Prevention, Atlanta, GA, United States
| |
Collapse
|
31
|
Guerrero D, Cantaert T, Missé D. Aedes Mosquito Salivary Components and Their Effect on the Immune Response to Arboviruses. Front Cell Infect Microbiol 2020; 10:407. [PMID: 32850501 PMCID: PMC7426362 DOI: 10.3389/fcimb.2020.00407] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Vector-borne diseases are responsible for over a billion infections each year and nearly one million deaths. Mosquito-borne dengue virus, West Nile, Japanese encephalitis, Zika, Chikungunya, and Rift Valley Fever viruses constitute major public health problems in regions with high densities of arthropod vectors. During the initial step of the transmission cycle, vector, host, and virus converge at the bite site, where local immune cells interact with the vector's saliva. Hematophagous mosquito saliva is a mixture of bioactive components known to modulate vertebrate hemostasis, immunity, and inflammation during the insect's feeding process. The capacity of mosquito saliva to modulate the host immune response has been well-studied over the last few decades and has led to the consensus that the presence of saliva is linked to the enhancement of virus transmission, host susceptibility, disease progression, viremia levels, and mortality. We review some of the major aspects of the interactions between mosquito saliva and the host immune response that may be useful for future studies on the control of arboviruses.
Collapse
Affiliation(s)
- David Guerrero
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Dorothée Missé
- MIVEGEC, IRD, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
32
|
Li Z, Guerrero F, Pérez de León AA, Foil LD, Swale DR. Small-Molecule Inhibitors of Inward Rectifier Potassium (Kir) Channels Reduce Bloodmeal Feeding and Have Insecticidal Activity Against the Horn Fly (Diptera: Muscidae). JOURNAL OF MEDICAL ENTOMOLOGY 2020; 57:1131-1140. [PMID: 32006426 DOI: 10.1093/jme/tjaa015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Indexed: 06/10/2023]
Abstract
Bloodmeal feeding by the horn fly, Haematobia irritans (L.), is associated with reduced milk production and blood loss that ultimately prevents weight gain of calves and yearlings. Thus, blood feeding by H. irritans causes significant economic losses in several continents. As with other arthropods, resistance to the majority of commercialized insecticides reduces the efficacy of current control programs. Thus, innovative technologies and novel biochemical targets for horn fly control are needed. Salivary gland and Malpighian tubule function are critical for H. irritans survivorship as they drive bloodmeal acquisition and maintain ion- and fluid homeostasis during bloodmeal processing, respectively. Experiments were conducted to test the hypothesis that pharmacological modulation of H. irritans inward rectifier potassium (Kir) channels would preclude blood feeding and induce mortality by reducing the secretory activity of the salivary gland while simultaneously inducing Malpighian tubule failure. Experimental results clearly indicate structurally diverse Kir channel modulators reduce the secretory activity of the salivary gland by up to fivefold when compared to control and the reduced saliva secretion was highly correlated to a reduction in bloodmeal acquisition in adult flies. Furthermore, adult feeding on blood treated with Kir channel modulators resulted in significant mortality. In addition to validating the Kir channels of H. irritans as putative insecticide targets, the knowledge gained from this study could be applied to develop novel therapeutic technologies targeting salivary gland or Malpighian tubule function to reduce the economic burden of horn fly ectoparasitism on cattle health and production.
Collapse
Affiliation(s)
- Zhilin Li
- Department of Entomology, Louisiana State University AgCenter, Baton Rouge, LA
| | | | - Adalberto A Pérez de León
- Knipling-Bushland Livestock Insects Research Laboratory and Veterinary Pest Genomics Center, United States Department of Agriculture-Agricultural Research Service, Kerrville, TX
| | - Lane D Foil
- Department of Entomology, Louisiana State University AgCenter, Baton Rouge, LA
| | - Daniel R Swale
- Department of Entomology, Louisiana State University AgCenter, Baton Rouge, LA
| |
Collapse
|
33
|
Christofferson RC, Parker DM, Overgaard HJ, Hii J, Devine G, Wilcox BA, Nam VS, Abubakar S, Boyer S, Boonnak K, Whitehead SS, Huy R, Rithea L, Sochantha T, Wellems TE, Valenzuela JG, Manning JE. Current vector research challenges in the greater Mekong subregion for dengue, Malaria, and Other Vector-Borne Diseases: A report from a multisectoral workshop March 2019. PLoS Negl Trop Dis 2020; 14:e0008302. [PMID: 32730249 PMCID: PMC7392215 DOI: 10.1371/journal.pntd.0008302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Rebecca C. Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Daniel M. Parker
- University of California, Irvine, California, United States of America
| | | | | | - Gregor Devine
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bruce A. Wilcox
- ASEAN Institute for Health Development, Mahidol University, Nakhon Pathom, Thailand
| | - Vu Sinh Nam
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Sazaly Abubakar
- Tropical Infectious Diseases Research and Education Center, Kuala Lumpur, Malaysia
| | | | - Kobporn Boonnak
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Stephen S. Whitehead
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Rekol Huy
- National Center for Parasitology Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Leang Rithea
- National Center for Parasitology Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Tho Sochantha
- National Center for Parasitology Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Thomas E. Wellems
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jesus G. Valenzuela
- National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jessica E. Manning
- US National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
| |
Collapse
|
34
|
Manning JE, Oliveira F, Coutinho-Abreu IV, Herbert S, Meneses C, Kamhawi S, Baus HA, Han A, Czajkowski L, Rosas LA, Cervantes-Medina A, Athota R, Reed S, Mateja A, Hunsberger S, James E, Pleguezuelos O, Stoloff G, Valenzuela JG, Memoli MJ. Safety and immunogenicity of a mosquito saliva peptide-based vaccine: a randomised, placebo-controlled, double-blind, phase 1 trial. Lancet 2020; 395:1998-2007. [PMID: 32534628 PMCID: PMC9151349 DOI: 10.1016/s0140-6736(20)31048-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND In animal models, immunity to mosquito salivary proteins protects animals against mosquito-borne disease. These findings provide a rationale to vaccinate against mosquito saliva instead of the pathogen itself. To our knowledge, no vector salivary protein-based vaccine has been tested for safety and immunogenicity in humans. We aimed to assess the safety and immunogenicity of Anopheles gambiae saliva vaccine (AGS-v), a peptide-based vaccine derived from four A gambiae salivary proteins, in humans. METHODS In this randomised, placebo-controlled, double-blind, phase 1 trial, participants were enrolled at the National Institutes of Health Clinical Center in Bethesda, MD, USA. Participants were eligible if they were healthy adults, aged 18-50 years with no history of severe allergic reactions to mosquito bites. Participants were randomly assigned (1:1:1), using block randomisation and a computer-generated randomisation sequence, to treatment with either 200 nmol of AGS-v vaccine alone, 200 nmol of AGS-v with adjuvant (Montanide ISA 51), or sterile water as placebo. Participants and clinicians were masked to treatment assignment. Participants were given a subcutaneous injection of their allocated treatment at day 0 and day 21, followed by exposure to feeding by an uninfected Aedes aegypti mosquito at day 42 to assess subsequent risk to mosquito bites in a controlled setting. The primary endpoints were safety and immunogenicity at day 42 after the first immunisation. Participants who were given at least one dose of assigned treatment were assessed for the primary endpoints and analysis was by intention to treat. The trial was registered with ClinicalTrials.gov, NCT03055000, and is closed for accrual. FINDINGS Between Feb 15 and Sept 10, 2017, we enrolled and randomly assigned 49 healthy adult participants to the adjuvanted vaccine (n=17), vaccine alone (n=16), or placebo group (n=16). Five participants did not complete the two-injection regimen with mosquito feeding at day 42, but were included in the safety analyses. No systemic safety concerns were identified; however, one participant in the adjuvanted vaccine group developed a grade 3 erythematous rash at the injection site. Pain, swelling, erythema, and itching were the most commonly reported local symptoms and were significantly increased in the adjuvanted vaccine group compared with both other treatment groups (nine [53%] of 17 participants in the adjuvanted vaccine group, two [13%] of 16 in the vaccine only group, and one [6%] of 16 in the placebo group; p=0·004). By day 42, participants who were given the adjuvanted vaccine had a significant increase in vaccine-specific total IgG antibodies compared with at baseline than did participants who were give vaccine only (absolute difference of log10-fold change of 0·64 [95% CI 0·39 to 0·89]; p=0·0002) and who were given placebo (0·62 [0·34 to 0·91]; p=0·0001). We saw a significant increase in IFN-γ production by peripheral blood mononuclear cells at day 42 in the adjuvanted vaccine group compared with in the placebo group (absolute difference of log10 ratio of vaccine peptide-stimulated vs negative control 0·17 [95% CI 0·061 to 0·27]; p=0·009) but we saw no difference between the IFN-γ production in the vaccine only group compared with the placebo group (0·022 [-0·072 to 0·116]; p=0·63). INTERPRETATION AGS-v was well tolerated, and, when adjuvanted, immunogenic. These findings suggest that vector-targeted vaccine administration in humans is safe and could be a viable option for the increasing burden of vector-borne disease. FUNDING Office of the Director and the Division of Intramural Research at the National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA.
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | | | - Samantha Herbert
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Claudio Meneses
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Holly Ann Baus
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Cervantes-Medina
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rani Athota
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Susan Reed
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, sponsored by the National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Sally Hunsberger
- Biostatistics Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Wang Y, Marin-Lopez A, Jiang J, Ledizet M, Fikrig E. Vaccination with Aedes aegypti AgBR1 Delays Lethal Mosquito-Borne Zika Virus Infection in Mice. Vaccines (Basel) 2020; 8:vaccines8020145. [PMID: 32218189 PMCID: PMC7348886 DOI: 10.3390/vaccines8020145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022] Open
Abstract
Zika Virus (ZIKV) is transmitted primarily by Aedes aegypti mosquitoes, resulting in asymptomatic infection, or acute illness with a fever and headache, or neurological complications, such as Guillain-Barre syndrome or fetal microcephaly. Previously, we determined that AgBR1, a mosquito salivary protein, induces inflammatory responses at the bite site, and that passive immunization with AgBR1 antiserum influences mosquito-transmitted ZIKV infection. Here, we show that the active immunization of mice with AgBR1 adjuvanted with aluminum hydroxide delays lethal mosquito-borne ZIKV infection, suggesting that AgBR1 may be used as part of a vaccine to combat ZIKV.
Collapse
Affiliation(s)
- Yuchen Wang
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan 430072, China
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Correspondence: or (Y.W.); (A.M.-L.)
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Correspondence: or (Y.W.); (A.M.-L.)
| | - Junjun Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- School of Public Health, Guangxi Medical University, Nanning 530021, China
| | | | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
36
|
Opasawatchai A, Yolwong W, Thuncharoen W, Inrueangsri N, Itsaradisaikul S, Sasisakulporn C, Jotikasthira W, Matangkasombut O, Reamtong O, Manuyakorn W, Songnuan W, Matangkasombut P. Novel salivary gland allergens from tropical mosquito species and IgE reactivity in allergic patients. World Allergy Organ J 2020; 13:100099. [PMID: 32099589 PMCID: PMC7031643 DOI: 10.1016/j.waojou.2020.100099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 12/29/2019] [Accepted: 01/15/2020] [Indexed: 11/16/2022] Open
Abstract
Background Mosquito allergy is common in tropical countries but remains under-diagnosed. This may be due to the lack of knowledge and diagnostic tools for tropical mosquito allergens. Objective We aimed to characterize allergens from tropical mosquito species and investigate IgE reactivity in mosquito-allergic patients to the salivary gland proteins from these mosquitoes. Methods Salivary gland extract (SGE) from 4 mosquito species, highly distributed in the tropics, including Aedes aegypti, Aedes albopictus, Culex quinquefasciatus, and Anopheles dirus b, were studied. SGE-specific IgE and IgG ELISA were developed, and serum from 64 mosquito-allergic and 22 non-allergic healthy control subjects was assayed. Further investigations using IgE-immunoblots followed by mass spectrometry analysis were performed to identify and characterize allergens from each species. Results Mosquito-allergic subjects have detectable serum IgE to SGE derived from local mosquito species, while the IgE levels to Aedes communis using commercially available ELISA were mostly minimal. IgE-immunoblot analysis and mass spectrometry identified 5 novel mosquito allergens from A. albopictus (Aed al 2, Aed al 3), C. quinquefasciatus (Cul q 2.01, Cul q 3), and A. dirus b (Ano d 2). Interestingly, 4 of the 5 new allergens belong to the D7 protein family. Conclusions & clinical relevance Five novel allergens from 3 tropical mosquito species were characterized. The majority of mosquito-allergic subjects who live in the tropics have IgE reactivity to these allergens. Our study paves the way for the development of diagnostic tests, component-resolved diagnostics, and future immunotherapy for mosquito allergy in tropical countries.
Collapse
Affiliation(s)
- Anunya Opasawatchai
- Department of Microbiology, Faculty of Science, Mahidol University, Thailand.,Faculty of Dentistry, Mahidol University, Thailand
| | | | | | | | - Sulak Itsaradisaikul
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Cherapat Sasisakulporn
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Wanlapa Jotikasthira
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Oranart Matangkasombut
- Department of Microbiology and Research Unit on Oral Microbiology and Immunology, Faculty of Dentistry, Chulalongkorn University, Thailand.,Laboratory of Biotechnology, Chulabhorn Research Institute, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Wiparat Manuyakorn
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand
| | - Wisuwat Songnuan
- Department of Plant Science, Faculty of Science, Mahidol University, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ponpan Matangkasombut
- Department of Microbiology, Faculty of Science, Mahidol University, Thailand.,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
37
|
Cabral S, de Paula A, Samuels R, da Fonseca R, Gomes S, Silva JR, Mury F. Aedes aegypti (Diptera: Culicidae) Immune Responses with Different Feeding Regimes Following Infection by the Entomopathogenic Fungus Metarhizium anisopliae. INSECTS 2020; 11:E95. [PMID: 32024202 PMCID: PMC7074208 DOI: 10.3390/insects11020095] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
Abstract
The mosquito Aedes aegypti is the most notorious vector of illness-causing viruses. The use of entomopathogenic fungi as bioinsecticides is a promising alternative for the development of novel mosquito control strategies. We investigate whether differences in immune responses could be responsible for modifications in survival rates of insects following different feeding regimes. Sucrose and blood-fed adult A. aegypti females were sprayed with M. anisopliae 1 × 106 conidia mL-1, and after 48 h, the midgut and fat body were dissected. We used RT-qPCR to monitor the expression of Cactus and REL1 (Toll pathway), IMD, REL2, and Caspar (IMD pathway), STAT and PIAS (JAK-STAT pathway), as well as the expression of antimicrobial peptides (Defensin A, Attacin and Cecropin G). REL1 and REL2 expression in both the midgut and fat body were higher in blood-fed fungus-challenged A. aegypti than in sucrose-fed counterparts. Interestingly, infection of sucrose-fed insects induced Cactus expression in the fat body, a negative regulator of the Toll pathway. The IMD gene was upregulated in the fat body in response to fungal infection after a blood meal. Additionally, we observed the induction of antimicrobial peptides in the blood-fed fungus-challenged insects. This study suggests that blood-fed A. aegypti are less susceptible to fungal infection due to the rapid induction of Toll and IMD immune pathways.
Collapse
Affiliation(s)
- Sara Cabral
- Laboratório Integrado de Bioquímica—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil; (S.C.); (J.R.S.)
| | - Adriano de Paula
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - Richard Samuels
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - Rodrigo da Fonseca
- Laboratório Integrado de Ciências Morfofuncionais—Instituto de Biodiversidade e Sustentabilidade –NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
| | - Simone Gomes
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - José Roberto Silva
- Laboratório Integrado de Bioquímica—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil; (S.C.); (J.R.S.)
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
| | - Flávia Mury
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
- Laboratório Integrado de Biociências Translacionais—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil
| |
Collapse
|
38
|
Montiel J, Carbal LF, Tobón-Castaño A, Vásquez GM, Fisher ML, Londono-Rentería B. IgG antibody response against Anopheles salivary gland proteins in asymptomatic Plasmodium infections in Narino, Colombia. Malar J 2020; 19:42. [PMID: 31973737 PMCID: PMC6979332 DOI: 10.1186/s12936-020-3128-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
Background The humoral immune response against Anopheles salivary glands proteins in the vertebrate host can reflect the intensity of exposure to Anopheles bites and the risk of Plasmodium infection. In Colombia, the identification of exposure biomarkers is necessary due to the several Anopheles species circulating. The purpose of this study was to evaluate risk of malaria infection by measuring antibody responses against salivary glands extracts from Anopheles (Nyssorhynchus) albimanus and Anopheles (Nys.) darlingi and also against the gSG6-P1 peptide of Anopheles gambiae in people residing in a malaria endemic area in the Colombian Pacific coast. Methods Dried blood spots samples were eluted to measure the IgG antibodies against salivary gland extracts of An. albimanus strains STECLA (STE) and Cartagena (CTG) and An. darlingi and the gSG6-P1 peptide by ELISA in uninfected people and microscopic and submicroscopic Plasmodium carriers from the Colombia Pacific Coast. A multiple linear mixed regression model, Spearman correlation, and Mann–Whitney U-test were used to analyse IgG data. Results Significant differences in specific IgG levels were detected between infected and uninfected groups for salivary glands extracts from An. albimanus and for gSG6-P1, also IgG response to CTG and gSG6-P1 peptide were positively associated with the IgG response to Plasmodium falciparum in the mixed model. Conclusion The CTG and STE An. albimanus salivary glands extracts are a potential source of new Anopheles salivary biomarkers to identify exposure to the main malaria vector and to calculate risk of disease in the Colombian Pacific coast. Also, the gSG6-P1 peptide has the potential to quantify human exposure to the subgenus Anopheles vectors in the same area.
Collapse
Affiliation(s)
- Jehidys Montiel
- Grupo Malaria, Universidad de Antioquia, Medellín, Colombia.,Department of Entomology, Kansas State University, Manhattan, KS, USA
| | - Luisa F Carbal
- Grupo Malaria, Universidad de Antioquia, Medellín, Colombia
| | - Alberto Tobón-Castaño
- Grupo Malaria, Universidad de Antioquia, Medellín, Colombia.,Facultad de Medicina, Instituto de Investigaciones Medicas, Universidad de Antioquia, Medellín, Colombia
| | | | | | | |
Collapse
|
39
|
Morens DM, Folkers GK, Fauci AS. Eastern Equine Encephalitis Virus - Another Emergent Arbovirus in the United States. N Engl J Med 2019; 381:1989-1992. [PMID: 31747726 DOI: 10.1056/nejmp1914328] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- David M Morens
- From the Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Gregory K Folkers
- From the Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Anthony S Fauci
- From the Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
40
|
Kaiser JA, Barrett ADT. Twenty Years of Progress Toward West Nile Virus Vaccine Development. Viruses 2019; 11:E823. [PMID: 31491885 PMCID: PMC6784102 DOI: 10.3390/v11090823] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
Although West Nile virus (WNV) has been a prominent mosquito-transmitted infection in North America for twenty years, no human vaccine has been licensed. With a cumulative number of 24,714 neurological disease cases and 2314 deaths in the U.S. since 1999, plus a large outbreak in Europe in 2018 involving over 2000 human cases in 15 countries, a vaccine is essential to prevent continued morbidity, mortality, and economic burden. Currently, four veterinary vaccines are licensed, and six vaccines have progressed into clinical trials in humans. All four veterinary vaccines require multiple primary doses and annual boosters, but for a human vaccine to be protective and cost effective in the most vulnerable older age population, it is ideal that the vaccine be strongly immunogenic with only a single dose and without subsequent annual boosters. Of six human vaccine candidates, the two live, attenuated vaccines were the only ones that elicited strong immunity after a single dose. As none of these candidates have yet progressed beyond phase II clinical trials, development of new candidate vaccines and improvement of vaccination strategies remains an important area of research.
Collapse
Affiliation(s)
- Jaclyn A Kaiser
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
41
|
Nouzova M, Clifton ME, Noriega FG. Mosquito adaptations to hematophagia impact pathogen transmission. CURRENT OPINION IN INSECT SCIENCE 2019; 34:21-26. [PMID: 31247413 DOI: 10.1016/j.cois.2019.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/28/2019] [Accepted: 02/04/2019] [Indexed: 06/09/2023]
Abstract
Mosquito-borne diseases such as Dengue fever, Chikungunya, and Malaria are critical threats to public health in many parts of the world. Female mosquitoes have evolved multiple adaptive mechanisms to hematophagy, including the ability to efficiently draw and digest blood, as well as the ability to eliminate excess fluids and toxic by-products of blood digestion. Pathogenic agents enter the mosquito digestive tract with the blood meal and need to travel through the midgut and into the hemocele in order to reach the salivary glands and infect a new host. Pathogens need to adjust to these hostile gut, hemocele, and salivary gland environments, and when possible influence the physiology and behavior of their hosts to enhance transmission.
Collapse
Affiliation(s)
- Marcela Nouzova
- Department of Biological Sciences and Biomolecular Science Institute, Florida International University, Miami, FL, USA; Institute of Parasitology, Biology Centre CAS, Ceske Budejovice, Czech Republic
| | - Mark E Clifton
- North Shore Mosquito Abatement District, Northfield, IL, USA
| | - Fernando G Noriega
- Department of Biological Sciences and Biomolecular Science Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
42
|
Ribeiro JM, Debat HJ, Boiani M, Ures X, Rocha S, Breijo M. An insight into the sialome, mialome and virome of the horn fly, Haematobia irritans. BMC Genomics 2019; 20:616. [PMID: 31357943 PMCID: PMC6664567 DOI: 10.1186/s12864-019-5984-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The horn fly (Haematobia irritans) is an obligate blood feeder that causes considerable economic losses in livestock industries worldwide. The control of this cattle pest is mainly based on insecticides; unfortunately, in many regions, horn flies have developed resistance. Vaccines or biological control have been proposed as alternative control methods, but the available information about the biology or physiology of this parasite is rather scarce. RESULTS We present a comprehensive description of the salivary and midgut transcriptomes of the horn fly (Haematobia irritans), using deep sequencing achieved by the Illumina protocol, as well as exploring the virome of this fly. Comparison of the two transcriptomes allow for identification of uniquely salivary or uniquely midgut transcripts, as identified by statistically differential transcript expression at a level of 16 x or more. In addition, we provide genomic highlights and phylogenetic insights of Haematobia irritans Nora virus and present evidence of a novel densovirus, both associated to midgut libraries of H. irritans. CONCLUSIONS We provide a catalog of protein sequences associated with the salivary glands and midgut of the horn fly that will be useful for vaccine design. Additionally, we discover two midgut-associated viruses that infect these flies in nature. Future studies should address the prevalence, biological effects and life cycles of these viruses, which could eventually lead to translational work oriented to the control of this economically important cattle pest.
Collapse
Affiliation(s)
- J. M. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway Room 3E28, Rockville, MD 20852 USA
| | - Humberto Julio Debat
- Instituto de Patología Vegetal, Centro de Investigaciones Agropecuarias, Instituto Nacional de Tecnología Agropecuaria (IPAVE-CIAP-INTA), Córdoba, Argentina
| | - M. Boiani
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125 Montevideo, Uruguay
| | - X. Ures
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125 Montevideo, Uruguay
| | - S. Rocha
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125 Montevideo, Uruguay
| | - M. Breijo
- Unidad de Reactivos y Biomodelos de Experimentación, Facultad de Medicina, Universidad de la República, Gral. Flores, 2125 Montevideo, Uruguay
| |
Collapse
|
43
|
Scarpassa VM, Debat HJ, Alencar RB, Saraiva JF, Calvo E, Arcà B, Ribeiro JMC. An insight into the sialotranscriptome and virome of Amazonian anophelines. BMC Genomics 2019; 20:166. [PMID: 30832587 PMCID: PMC6399984 DOI: 10.1186/s12864-019-5545-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/18/2019] [Indexed: 01/17/2023] Open
Abstract
Background Saliva of mosquitoes contains anti-platelet, anti-clotting, vasodilatory, anti-complement and anti-inflammatory substances that help the blood feeding process. The salivary polypeptides are at a fast pace of evolution possibly due to their relative lack of structural constraint and possibly also by positive selection on their genes leading to evasion of host immune pressure. Results In this study, we used deep mRNA sequence to uncover for the first time the sialomes of four Amazonian anophelines species (Anopheles braziliensis, A. marajorara, A. nuneztovari and A. triannulatus) and extend the knowledge of the A. darlingi sialome. Two libraries were generated from A. darlingi mosquitoes, sampled from two localities separated ~ 1100 km apart. A total of 60,016 sequences were submitted to GenBank, which will help discovery of novel pharmacologically active polypeptides and the design of specific immunological markers of mosquito exposure. Additionally, in these analyses we identified and characterized novel phasmaviruses and anpheviruses associated to the sialomes of A. triannulatus, A. marajorara and A. darlingi species. Conclusions Besides their pharmacological properties, which may be exploited for the development of new drugs (e.g. anti-thrombotics), salivary proteins of blood feeding arthropods may be turned into tools to prevent and/or better control vector borne diseases; for example, through the development of vaccines or biomarkers to evaluate human exposure to vector bites. The sialotranscriptome study reported here provided novel data on four New World anopheline species and allowed to extend our knowledge on the salivary repertoire of A. darlingi. Additionally, we discovered novel viruses following analysis of the transcriptomes, a procedure that should become standard within future RNAseq studies. Electronic supplementary material The online version of this article (10.1186/s12864-019-5545-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vera Margarete Scarpassa
- Laboratório de Genética de Populações e Evolução de Mosquitos Vetores de Malária e Dengue, Coordenação de Biodiversidade, Instituto Nacional de Pesquisas da Amazônia, Manaus, Amazonas, Brazil
| | - Humbeto Julio Debat
- Instituto de Patología Vegetal, Centro de Investigaciones Agropecuarias, Instituto Nacional de Tecnología Agropecuaria (IPAVE-CIAP-INTA), Córdoba, Argentina
| | - Ronildo Baiatone Alencar
- Laboratório de Genética de Populações e Evolução de Mosquitos Vetores de Malária e Dengue, Coordenação de Biodiversidade, Instituto Nacional de Pesquisas da Amazônia, Manaus, Amazonas, Brazil
| | - José Ferreira Saraiva
- Laboratório de Genética de Populações e Evolução de Mosquitos Vetores de Malária e Dengue, Coordenação de Biodiversidade, Instituto Nacional de Pesquisas da Amazônia, Manaus, Amazonas, Brazil
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University of Rome, Rome, Italy
| | - José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
| |
Collapse
|
44
|
Arcà B, Colantoni A, Fiorillo C, Severini F, Benes V, Di Luca M, Calogero RA, Lombardo F. MicroRNAs from saliva of anopheline mosquitoes mimic human endogenous miRNAs and may contribute to vector-host-pathogen interactions. Sci Rep 2019; 9:2955. [PMID: 30814633 PMCID: PMC6393464 DOI: 10.1038/s41598-019-39880-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/04/2019] [Indexed: 12/31/2022] Open
Abstract
During blood feeding haematophagous arthropods inject into their hosts a cocktail of salivary proteins whose main role is to counteract host haemostasis, inflammation and immunity. However, animal body fluids are known to also carry miRNAs. To get insights into saliva and salivary gland miRNA repertoires of the African malaria vector Anopheles coluzzii we used small RNA-Seq and identified 214 miRNAs, including tissue-enriched, sex-biased and putative novel anopheline miRNAs. Noteworthy, miRNAs were asymmetrically distributed between saliva and salivary glands, suggesting that selected miRNAs may be preferentially directed toward mosquito saliva. The evolutionary conservation of a subset of saliva miRNAs in Anopheles and Aedes mosquitoes, and in the tick Ixodes ricinus, supports the idea of a non-random occurrence pointing to their possible physiological role in blood feeding by arthropods. Strikingly, eleven of the most abundant An. coluzzi saliva miRNAs mimicked human miRNAs. Prediction analysis and search for experimentally validated targets indicated that miRNAs from An. coluzzii saliva may act on host mRNAs involved in immune and inflammatory responses. Overall, this study raises the intriguing hypothesis that miRNAs injected into vertebrates with vector saliva may contribute to host manipulation with possible implication for vector-host interaction and pathogen transmission.
Collapse
Affiliation(s)
- Bruno Arcà
- Department of Public Health and Infectious Diseases, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Alessio Colantoni
- Department of Biology and Biotechnology, "Sapienza University", Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Carmine Fiorillo
- Department of Public Health and Infectious Diseases, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Francesco Severini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Marco Di Luca
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Raffaele A Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases, "Sapienza" University, Piazzale Aldo Moro 5, 00185, Rome, Italy
| |
Collapse
|
45
|
Manning JE, Cantaert T. Time to Micromanage the Pathogen-Host-Vector Interface: Considerations for Vaccine Development. Vaccines (Basel) 2019; 7:E10. [PMID: 30669682 PMCID: PMC6466432 DOI: 10.3390/vaccines7010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The current increase in vector-borne disease worldwide necessitates novel approaches to vaccine development targeted to pathogens delivered by blood-feeding arthropod vectors into the host skin. A concept that is gaining traction in recent years is the contribution of the vector or vector-derived components, like salivary proteins, to host-pathogen interactions. Indeed, the triad of vector-host-pathogen interactions in the skin microenvironment can influence host innate and adaptive responses alike, providing an advantage to the pathogen to establish infection. A better understanding of this "bite site" microenvironment, along with how host and vector local microbiomes immunomodulate responses to pathogens, is required for future vaccines for vector-borne diseases. Microneedle administration of such vaccines may more closely mimic vector deposition of pathogen and saliva into the skin with the added benefit of near painless vaccine delivery. Focusing on the 'micro'⁻from microenvironments to microbiomes to microneedles⁻may yield an improved generation of vector-borne disease vaccines in today's increasingly complex world.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh 12201, Cambodia.
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12201, Cambodia.
| |
Collapse
|
46
|
Arcà B, Ribeiro JM. Saliva of hematophagous insects: a multifaceted toolkit. CURRENT OPINION IN INSECT SCIENCE 2018; 29:102-109. [PMID: 30551815 DOI: 10.1016/j.cois.2018.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 07/20/2018] [Indexed: 06/09/2023]
Abstract
Transcriptomic, proteomic and genomic studies significantly improved our understanding of the complexity of blood feeding insect saliva providing unparalleled evolutionary insights. Salivary genes appeared to be under strong selective pressure with gene duplication and functional diversification being a powerful driver in the evolution of novel salivary genes/functions. The first insect salivary proteins responsible for complement inhibition were identified and a widespread mechanism of action shared by unrelated salivary protein families was recognized and named kratagonism. microRNAs were for the first time described in the saliva of a few blood feeding arthropods raising intriguing questions on their possible contribution to vertebrate host manipulation and pathogen transmission and further emphasizing how much we still have to learn on blood feeding insect saliva.
Collapse
Affiliation(s)
- Bruno Arcà
- Department of Public Health and Infectious Diseases, "Sapienza" University, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Josè Mc Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| |
Collapse
|
47
|
Bakhshi H, Failloux AB, Zakeri S, Raz A, Dinparast Djadid N. Mosquito-borne viral diseases and potential transmission blocking vaccine candidates. INFECTION GENETICS AND EVOLUTION 2018; 63:195-203. [PMID: 29842982 DOI: 10.1016/j.meegid.2018.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 01/17/2023]
Abstract
Mosquito-borne viral diseases (MBVDs) have a complex biological cycle involving vectors and vertebrate hosts. These viruses are responsible for many deadly diseases worldwide. Although MBVDs threaten mostly developing countries, there is growing evidence indicating that they are also of concern in western countries where local transmission of arboviruses such as West Nile, Zika, Chikungunya and Dengue viruses have been recently reported. The rapid rise in human infections caused by these viruses is attributed to rapid climate change and travel facilities. Usually, the only way to control these diseases relies on the control of vectors in the absence of licensed vaccines and specific treatments. However, the overuse of insecticides has led to the emergence of insecticide resistance in vector populations, posing significant challenges for their control. An alternative method for reducing MBVDs can be the use of Transmission Blocking Vaccines (TBVs) that limits viral infection at the mosquito vector stage. Some successes have been obtained confirming the potential application of TBVs against viruses; however, this approach remains at the developmental stage and still needs improvements. The present review aims to give an update on MBVDs and to discuss the application as well as usage of potential TBVs for the control of mosquito-borne viral infections.
Collapse
Affiliation(s)
- Hasan Bakhshi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Tehran, Iran
| | - Anna-Bella Failloux
- Department of Virology, Arboviruses and Insect Vectors, Institut Pasteur, Paris, France
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Tehran, Iran.
| |
Collapse
|