1
|
Hall RA, Nguyen W, Khromykh AA, Suhrbier A. Insect-specific virus platforms for arbovirus vaccine development. Front Immunol 2025; 16:1521104. [PMID: 40160816 PMCID: PMC11949993 DOI: 10.3389/fimmu.2025.1521104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Certain insect-specific viruses (ISVs), specifically the mosquito alphaviruses, Eilat and Yada Yada viruses, and orthoflaviviruses, Binjari, Aripo, YN15-283-02 and Chaoyang viruses, have emerged as potential platforms for generation of whole virus vaccines for human and veterinary applications. These ISVs are remarkably tolerant of the substitution of their structural polyproteins with those of alphaviruses and orthoflaviviruses that are pathogenic in humans and/or animals. The resulting ISV-based chimeric vaccines have been evaluated in mouse models and have demonstrated safety and efficacy in non-human primates, crocodiles and pigs. Targets include chikungunya, Venezuelan and eastern equine encephalitis, dengue, Zika, yellow fever, Japanese encephalitis and West Nile viruses. ISV-based chimeric vaccines provide authentically folded tertiary and quaternary whole virion particle structures to the immune system, a key feature for induction of protective antibody responses. These vaccines are manufactured in C6/36 or C7-10 mosquito cell lines, where they grow to high titers, but they do not replicate in vertebrate vaccine recipients. This review discusses the progress of these emerging technologies and addresses challenges related to adjuvanting, safety, and manufacturing.
Collapse
Affiliation(s)
- Roy A. Hall
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD, Australia
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD, Australia
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
| | - Andreas Suhrbier
- Global Virus Network Centre of Excellence, Australian Infectious Diseases Research Centre, Brisbane, QLD, Australia
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
2
|
Abdeladhim M, Teixeira C, Ressner R, Hummer K, Dey R, Gomes R, de Castro W, de Araujo FF, Turiansky GW, Iniguez E, Meneses C, Oliveira F, Aronson N, Lacsina JR, Valenzuela JG, Kamhawi S. Lutzomyia longipalpis salivary proteins elicit human innate and adaptive immune responses detrimental to Leishmania parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640210. [PMID: 40196468 PMCID: PMC11974753 DOI: 10.1101/2025.02.25.640210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Leishmania parasites are transmitted via the bite of infected sand flies, whose saliva modulates host immune responses to promote Leishmania infection, especially in unexposed individuals. For humans in endemic areas, the immune consequences of chronic exposure to sand fly saliva remain poorly understood. We performed a human challenge study with Lutzomyia longipalpis, the primary vector of visceral leishmaniasis in the Americas. Fifteen healthy volunteers were exposed multiple times to uninfected Lu. longipalpis bites over the course of a year. PBMCs collected after several exposures were stimulated ex vivo by recombinant Lu. longipalpis salivary proteins to measure cytokine responses. Two salivary proteins, LJM19 and LJL143, elicited TH1-polarized cytokine responses, but with high co-expression of the TH2 cytokine IL-13. LJM19 also induced higher levels of IL-6 and IL-7, while both LJM19 and LJL143 induced the innate cytokines IL-1β and IFN-α. Importantly, TH1 polarization induced by LJM19 or LJL143 in PBMCs correlated with enhanced killing of Leishmania in co-cultured macrophages. Skin biopsies from two volunteers revealed bite site infiltration with CD4-CD8- T cells. Our data suggest that sand fly exposed individuals demonstrate robust innate and adaptive cellular immune responses to vector salivary proteins that can be co-opted to protect humans against Leishmania infection.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Clarissa Teixeira
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Roseanne Ressner
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States
| | - Kelly Hummer
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States
| | - Regis Gomes
- Laboratory of Immunoparasitology, Department of Biotechnology, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Waldionê de Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fernanda Fortes de Araujo
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States
| | - George W. Turiansky
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Naomi Aronson
- Walter Reed National Military Medical Center, Bethesda, Maryland, United States
- Infectious Diseases Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States
| | - Joshua R. Lacsina
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States
| |
Collapse
|
3
|
Goldman OV, DeFoe AE, Qi Y, Jiao Y, Weng SC, Houri-Zeevi L, Lakhiani P, Morita T, Razzauti J, Rosas-Villegas A, Tsitohay YN, Walker MM, Hopkins BR, Akbari OS, Duvall LB, White-Cooper H, Sorrells TR, Sharma R, Li H, Vosshall LB, Shai N. Mosquito Cell Atlas: A single-nucleus transcriptomic atlas of the adult Aedes aegypti mosquito. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.639765. [PMID: 40060408 PMCID: PMC11888250 DOI: 10.1101/2025.02.25.639765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The female mosquito's remarkable ability to hunt humans and transmit pathogens relies on her unique biology. Here, we present the Mosquito Cell Atlas (MCA), a comprehensive single-nucleus RNA sequencing dataset of more than 367,000 nuclei from 19 dissected tissues of adult female and male Aedes aegypti, providing cellular-level resolution of mosquito biology. We identify novel cell types and expand our understanding of sensory neuron organization of chemoreceptors to all sensory tissues. Our analysis uncovers male-specific cells and sexually dimorphic gene expression in the antenna and brain. In female mosquitoes, we find that glial cells in the brain, rather than neurons, undergo the most extensive transcriptional changes following blood feeding. Our findings provide insights into the cellular basis of mosquito behavior and sexual dimorphism. The MCA aims to serve as a resource for the vector biology community, enabling systematic investigation of cell-type specific expression across all mosquito tissues.
Collapse
Affiliation(s)
- Olivia V. Goldman
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| | - Alexandra E. DeFoe
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yaoyu Jiao
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shih-Che Weng
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Leah Houri-Zeevi
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Priyanka Lakhiani
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Takeshi Morita
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Jacopo Razzauti
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Price Family Center for the Social Brain, The Rockefeller University, New York, NY 10065, USA
| | - Adriana Rosas-Villegas
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Yael N. Tsitohay
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Madison M. Walker
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Ben R. Hopkins
- Department of Evolution and Ecology, University of California Davis, Davis, CA 95616, USA
| | | | - Omar S. Akbari
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laura B. Duvall
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Helen White-Cooper
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Trevor R. Sorrells
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
- Howard Hughes Medical Institute, New Haven, CT 06510, USA
| | - Roshan Sharma
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Single-cell Analytics Innovation Lab, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leslie B. Vosshall
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Nadav Shai
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| |
Collapse
|
4
|
Yui S, Fujii N, Terauchi J, Tanabe N, Kanno M, Umehara K, Iijima R, Kamata R, Ohkura N, Kishimoto S, Sasaki T. Chemotactic Activity of Products of Elizabethkingia anophelis Derived from Aedes albopictus against RAW264 Murine Macrophage Cell Line. Jpn J Infect Dis 2025; 78:35-42. [PMID: 39477519 DOI: 10.7883/yoken.jjid.2024.227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Dengue viruses enter the dermal macrophages derived from other tissues following a bite from an infected mosquito. We examined the chemotactic activity of factors derived from the dengue vector mosquito Aedes albopictus on a RAW264 murine macrophage cell line. We found that Elizabethkingia anophelis isolated from the mosquitoes exhibits migration-inducing activity in RAW264 cells. The active substances that induce the chemotactic movement were extracted using ethyl acetate. Chemotactic activity was noted in several of the fractions isolated using reverse-phase chromatography, suggesting that multiple components were responsible for this activity. Next, we isolated three bacterial colonies from wild A. albopictus mosquitoes collected from Toyama Park (Tokyo, Japan). The bacterial 16S rRNA gene sequences shared homology with that of Lonsdalea quercina. These bacteria also exhibited migration-inducing activity in RAW264 cells. The migration-inducing activity of the bacteria in mosquitoes may be a novel aspect of mosquito-mediated viral infections.
Collapse
Affiliation(s)
- Satoru Yui
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | | | - Jo Terauchi
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Nana Tanabe
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Marie Kanno
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Kouta Umehara
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Ryosuke Iijima
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Riyo Kamata
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | - Naoki Ohkura
- Department of Pharmaceutical Sciences, Teikyo University, Japan
| | | | - Toshinori Sasaki
- Department of Medical Entomology, National Institute of Infectious Diseases, Japan
| |
Collapse
|
5
|
Guo J, He X, Tao J, Sun H, Yang J. Unraveling the Molecular Mechanisms of Mosquito Salivary Proteins: New Frontiers in Disease Transmission and Control. Biomolecules 2025; 15:82. [PMID: 39858476 PMCID: PMC11764250 DOI: 10.3390/biom15010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/13/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Mosquito-borne diseases are a group of illnesses caused by pathogens transmitted by mosquitoes, and they are globally prevalent, particularly in tropical and subtropical regions. Pathogen transmission occurs during mosquito blood feeding, a process in which mosquito saliva plays a crucial role. Mosquito saliva contains a variety of biologically active proteins that facilitate blood feeding by preventing blood clotting, promoting vasodilation, and modulating the host's immune and inflammatory responses. These effects create an environment conducive to pathogen invasion and dissemination. Specific mosquito salivary proteins (MSPs) can promote pathogen transmission through mechanisms that either regulate hosts' anti-infective immune responses or directly enhance pathogens' activity. Strategies targeting these MSPs have emerged as an innovative and promising approach for the control of mosquito-borne diseases. Meanwhile, the diversity of these proteins and their complex interactions with the host immune system necessitate further research to develop safer and more effective interventions. This review examines the functional diversity of MSPs and their roles in disease transmission, discusses the advantages and challenges of strategies targeting these proteins, and explores potential future directions for research in this area.
Collapse
Affiliation(s)
- Jiayin Guo
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (J.G.); (X.H.); (H.S.)
| | - Xiaoe He
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (J.G.); (X.H.); (H.S.)
| | - Jianli Tao
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Hui Sun
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (J.G.); (X.H.); (H.S.)
| | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (J.G.); (X.H.); (H.S.)
| |
Collapse
|
6
|
Cecilia H, Althouse BM, Azar SR, Moehn BA, Yun R, Rossi SL, Vasilakis N, Hanley KA. Aedes albopictus is not an arbovirus aficionado when feeding on cynomolgus macaques or squirrel monkeys. iScience 2024; 27:111198. [PMID: 39555418 PMCID: PMC11563999 DOI: 10.1016/j.isci.2024.111198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/27/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Viruses transmitted by Aedes mosquitoes (e.g., dengue [DENV], Zika [ZIKV]) have demonstrated high potential to spill over from their ancestral, sylvatic cycles in non-human primates to establish transmission in humans. Epidemiological models require accurate knowledge of the contact structure between hosts and vectors, which is highly sensitive to any impacts of virus infection in mosquitoes or hosts on mosquito feeding behavior. Current evidence for whether these viruses affect vector behavior is mixed. Here we leveraged a study on sylvatic DENV-2 and ZIKV transmission between two species of monkey and Aedes albopictus to determine whether virus infection of either host or vector alters vector feeding behavior. Engorgement rates varied from 0% to 100%, but this was not driven by vector nor host infection, but rather by the individual host, host species, and host body temperature. 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, USA
| | - 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
|
7
|
Pyuza JJ, van Dorst MM, Stam K, Wammes L, König M, Kullaya VI, Kruize Y, Huisman W, Andongolile N, Ngowi A, Shao ER, Mremi A, Hogendoorn PC, Msuya SE, Jochems SP, de Steenhuijsen Piters WA, Yazdanbakhsh M. Lifestyle score is associated with cellular immune profiles in healthy Tanzanian adults. Brain Behav Immun Health 2024; 41:100863. [PMID: 39398291 PMCID: PMC11470418 DOI: 10.1016/j.bbih.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Immune system and vaccine responses vary across geographical locations worldwide, not only between high and low-middle income countries (LMICs), but also between rural and urban populations within the same country. Lifestyle factors such as housing conditions, exposure to microorganisms and parasites and diet are associated with rural-and urban-living. However, the relationships between these lifestyle factors and immune profiles have not been mapped in detail. Here, we profiled the immune system of 100 healthy Tanzanians living across four rural/urban areas using mass cytometry. We developed a lifestyle score based on an individual's household assets, housing condition and recent dietary history and studied the association with cellular immune profiles. Seventeen out of 80 immune cell clusters were associated with living location or lifestyle score, with eight identifiable only using lifestyle score. Individuals with low lifestyle score, most of whom live in rural settings, showed higher frequencies of NK cells, plasmablasts, atypical memory B cells, T helper 2 cells, regulatory T cells and activated CD4+ T effector memory cells expressing CD38, HLA-DR and CTLA-4. In contrast, those with high lifestyle score, most of whom live in urban areas, showed a less activated state of the immune system illustrated by higher frequencies of naïve CD8+ T cells. Using an elastic net machine learning model, we identified cellular immune signatures most associated with lifestyle score. Assuming a link between these immune profiles and vaccine responses, these signatures may inform us on the cellular mechanisms underlying poor responses to vaccines, but also reduced autoimmunity and allergies in low- and middle-income countries.
Collapse
Affiliation(s)
- Jeremia J. Pyuza
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Marloes M.A.R. van Dorst
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Koen Stam
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Linda Wammes
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Marion König
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Vesla I. Kullaya
- Kilimanjaro Clinical Research Institute (KCRI), Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Department of Medical Biochemistry and Molecular Biology, Kilimanjaro Christian Medical College (KCMUCo), Moshi, Tanzania
| | - Yvonne Kruize
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Wesley Huisman
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | - Nikuntufya Andongolile
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Anastazia Ngowi
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Elichilia R. Shao
- Department of Internal Medicine, Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Alex Mremi
- Department of Pathology, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | | | - Sia E. Msuya
- Institute of Public Health, Kilimanjaro Christian University Medical College (KCMUCo), Moshi, Tanzania
- Department of Community Medicine, Kilimanjaro Christian Medical Centre (KCMC), Moshi, Tanzania
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| | | | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, ZA, Leiden, Netherlands
| |
Collapse
|
8
|
Bourne ME, Lucas-Barbosa D, Verhulst NO. Host location by arthropod vectors: are microorganisms in control? CURRENT OPINION IN INSECT SCIENCE 2024; 65:101239. [PMID: 39067510 DOI: 10.1016/j.cois.2024.101239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Vector-borne microorganisms are dependent on their arthropod vector for their transmission to and from vertebrates. The 'parasite manipulation hypothesis' states that microorganisms are likely to evolve manipulations of such interactions for their own selective benefit. Recent breakthroughs uncovered novel ecological interactions initiated by vector-borne microorganisms, which are linked to different stages of the host location by their arthropod vectors. Therefore, we give an actualised overview of the various means through which vector-borne microorganisms impact their vertebrate and arthropod hosts to ultimately benefit their own transmission. Harnessing the directionality and underlying mechanisms of these interactions driven by vector-borne microorganisms may provide tools to reduce the spread of pathogenic vector-borne microorganisms.
Collapse
Affiliation(s)
- Mitchel E Bourne
- National Centre for Vector Entomology, Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zürich, Winterthurerstrasse 266A, 8057 Zürich, Switzerland.
| | - Dani Lucas-Barbosa
- National Centre for Vector Entomology, Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zürich, Winterthurerstrasse 266A, 8057 Zürich, Switzerland; Research Institute of Organic Agriculture FiBL, Ackerstrasse 113, 5070 Frick, Switzerland
| | - Niels O Verhulst
- National Centre for Vector Entomology, Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zürich, Winterthurerstrasse 266A, 8057 Zürich, Switzerland.
| |
Collapse
|
9
|
Bursali F, Touray M. The complexities of blood-feeding patterns in mosquitoes and sandflies and the burden of disease: A minireview. Vet Med Sci 2024; 10:e1580. [PMID: 39171609 PMCID: PMC11339650 DOI: 10.1002/vms3.1580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Mosquitoes and sandflies exhibit a wide range of blood feeding patterns, targeting a wide range of vertebrate species, including birds, mammals, reptiles, and amphibians, for proteins vital for egg development. This broad host range increases the opportunity for them to acquire pathogens of numerous debilitating-and-fatal diseases from various animal reservoirs, playing a significant role in disease crossover between animals and humans, also known as zoonotic transmission. This review focuses on the intricate blood-feeding habits of these dipteran vectors, their sensory systems and the complex dance between host and pathogen during disease transmission. We delve into the influence of blood sources on pathogen spread by examining the insect immune response and its intricate interplay with pathogens. The remarkable sense of smell guiding them towards food sources and hosts is explored, highlighting the interplay of multiple sensory cues in their navigation. Finally, we examine the challenges in mosquito control strategies and explore innovations in this field, emphasizing the need for sustainable solutions to combat this global health threat. By understanding the biology and behaviour of these insects, we can develop more effective strategies to protect ourselves and mitigate the burden of vector-borne diseases.
Collapse
Affiliation(s)
- Fatma Bursali
- Biology Department, Faculty of ScienceAydin Adnan Menderes UniversityAydinTürkiye
| | - Mustapha Touray
- Biology Department, Faculty of ScienceAydin Adnan Menderes UniversityAydinTürkiye
| |
Collapse
|
10
|
Marin-Lopez A, Huck JD, Esterly AT, Azcutia V, Rosen C, Garcia-Milian R, Sefik E, Vidal-Pedrola G, Raduwan H, Chen TY, Arora G, Halene S, Shaw AC, Palm NW, Flavell RA, Parkos CA, Thangamani S, Ring AM, Fikrig E. The human CD47 checkpoint is targeted by an immunosuppressive Aedes aegypti salivary factor to enhance arboviral skin infectivity. Sci Immunol 2024; 9:eadk9872. [PMID: 39121194 PMCID: PMC11924945 DOI: 10.1126/sciimmunol.adk9872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 05/02/2024] [Accepted: 07/16/2024] [Indexed: 08/11/2024]
Abstract
The Aedes aegypti mosquito is a vector of many infectious agents, including flaviviruses such as Zika virus. Components of mosquito saliva have pleomorphic effects on the vertebrate host to enhance blood feeding, and these changes also create a favorable niche for pathogen replication and dissemination. Here, we demonstrate that human CD47, which is known to be involved in various immune processes, interacts with a 34-kilodalton mosquito salivary protein named Nest1. Nest1 is up-regulated in blood-fed female A. aegypti and facilitates Zika virus dissemination in human skin explants. Nest1 has a stronger affinity for CD47 than its natural ligand, signal regulatory protein α, competing for binding at the same interface. The interaction between Nest1 with CD47 suppresses phagocytosis by human macrophages and inhibits proinflammatory responses by white blood cells, thereby suppressing antiviral responses in the skin. This interaction elucidates how an arthropod protein alters the human response to promote arbovirus infectivity.
Collapse
Affiliation(s)
- Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John D Huck
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Allen T Esterly
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Connor Rosen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rolando Garcia-Milian
- Bioinformatics Support Program, Cushing/Whitney Medical Library, Yale School of Medicine, New Haven, CT, USA
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Gemma Vidal-Pedrola
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Saravanan Thangamani
- Department of Microbiology and Immunology, State University of New York, Upstate Medical University, Syracuse, NY, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
11
|
Blanken SL, Barry A, Lanke K, Guelbeogo M, Ouedraogo A, Soulama I, Coulibaly SA, Teelen K, Graumans W, Dumont E, Stone W, Ramjith J, Marti M, Andrade CM, Drakeley C, Collins K, Tiono A, Bousema T. Plasmodium falciparum gametocyte production correlates with genetic markers of parasite replication but is not influenced by experimental exposure to mosquito biting. EBioMedicine 2024; 105:105190. [PMID: 38901148 PMCID: PMC11239461 DOI: 10.1016/j.ebiom.2024.105190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Plasmodium blood-stage parasites balance asexual multiplication with gametocyte development. Few studies link these dynamics with parasite genetic markers in vivo; even fewer in longitudinally monitored infections. Environmental influences on gametocyte formation, such as mosquito exposure, may influence the parasite's investment in gametocyte production. METHODS We investigated gametocyte production and asexual multiplication in two Plasmodium falciparum infected populations; a controlled human malaria infection (CHMI) study and a 28-day observational study in naturally infected individuals in Burkina Faso with controlled mosquito exposure. We measured gene transcript levels previously related to gametocyte formation (ap2-g, surfin1.2, surfin13.1, gexp-2) or inhibition of asexual multiplication (sir2a) and compared transcript levels to ring-stage parasite and mature gametocyte densities. FINDINGS Three of the five markers (ap2-g, surfin1.2, surfin13.1) predicted peak gametocytaemia in the CHMI study. An increase in all five markers in natural infections was associated with an increase in mature gametocytes 14 days later; the effect of sir2a on future gametocytes was strongest (fold change = 1.65, IQR = 1.22-2.24, P = 0.004). Mosquito exposure was not associated with markers of gametocyte formation (ap2-g P = 0.277; sir2a P = 0.499) or carriage of mature gametocytes (P = 0.379). INTERPRETATION All five parasite genetic markers predicted gametocyte formation over a single cycle of gametocyte formation and maturation in vivo; sir2a and ap2-g were most closely associated with gametocyte growth dynamics. We observed no evidence to support the hypothesis that exposure to Anopheles mosquito bites stimulates gametocyte formation. FUNDING This work was funded by the Bill & Melinda Gates Foundation (INDIE OPP1173572), the European Research Council fellowship (ERC-CoG 864180) and UKRI Medical Research Council (MR/T016272/1) and Wellcome Center (218676/Z/19/Z).
Collapse
Affiliation(s)
- Sara Lynn Blanken
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Aissata Barry
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou, Burkina Faso
| | - Kjerstin Lanke
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Moussa Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Alphonse Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Issiaka Soulama
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Sam Aboubacar Coulibaly
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Karina Teelen
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Elin Dumont
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Will Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Jordache Ramjith
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection and Immunity, University of Glasgow, Glasgow, Scotland, UK
| | - Carolina M Andrade
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Chris Drakeley
- MRC International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Katharine Collins
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Alfred Tiono
- Centre National de Recherche et de Formation sur le Paludisme (CNRFP), Ouagadougou 01, Burkina Faso
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
12
|
Wang ZY, Nie KX, Niu JC, Cheng G. Research progress toward the influence of mosquito salivary proteins on the transmission of mosquito-borne viruses. INSECT SCIENCE 2024; 31:663-673. [PMID: 37017683 DOI: 10.1111/1744-7917.13193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Mosquito-borne viruses (MBVs) are a large class of viruses transmitted mainly through mosquito bites, including dengue virus, Zika virus, Japanese encephalitis virus, West Nile virus, and chikungunya virus, which pose a major threat to the health of people around the world. With global warming and extended human activities, the incidence of many MBVs has increased significantly. Mosquito saliva contains a variety of bioactive protein components. These not only enable blood feeding but also play a crucial role in regulating local infection at the bite site and the remote dissemination of MBVs as well as in remodeling the innate and adaptive immune responses of host vertebrates. Here, we review the physiological functions of mosquito salivary proteins (MSPs) in detail, the influence and the underlying mechanism of MSPs on the transmission of MBVs, and the current progress and issues that urgently need to be addressed in the research and development of MSP-based MBV transmission blocking vaccines.
Collapse
Affiliation(s)
- Zhao-Yang Wang
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Kai-Xiao Nie
- Department of Pathogen Biology, School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ji-Chen Niu
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Gong Cheng
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Bevivino G, Maurizi L, Ammendolia MG, Longhi C, Arcà B, Lombardo F. Peptides with Antimicrobial Activity in the Saliva of the Malaria Vector Anopheles coluzzii. Int J Mol Sci 2024; 25:5529. [PMID: 38791567 PMCID: PMC11121840 DOI: 10.3390/ijms25105529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Mosquito saliva plays a crucial physiological role in both sugar and blood feeding by helping sugar digestion and exerting antihemostatic functions. During meal acquisition, mosquitoes are exposed to the internalization of external microbes. Since mosquitoes reingest significant amounts of saliva during feeding, we hypothesized that salivary antimicrobial components may participate in the protection of mouthparts, the crop, and the gut by inhibiting bacterial growth. To identify novel potential antimicrobials from mosquito saliva, we selected 11 candidates from Anopheles coluzzii salivary transcriptomic datasets and obtained them either using a cell-free transcription/translation expression system or, when feasible, via chemical synthesis. Hyp6.2 and hyp13, which were predicted to be produced as propeptides and cleaved in shorter mature forms, showed the most interesting results in bacterial growth inhibition assays. Hyp6.2 (putative mature form, 35 amino acid residues) significantly inhibited the growth of Gram-positive (Staphylococcus aureus) and Gram-negative (Escherichia coli and Serratia marcescens) bacteria. Hyp13 (short form, 19 amino acid residues) dose-dependently inhibited E. coli and S. marcescens growth, inducing membrane disruption in both Gram-positive and Gram-negative bacteria as indicated with scanning electron microscopy. In conclusion, we identified two A. coluzzii salivary peptides inhibiting Gram-positive and Gram-negative bacteria growth and possibly contributing to the protection of mosquito mouthparts and digestive tracts from microbial infection during and/or after feeding.
Collapse
Affiliation(s)
- Giulia Bevivino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Linda Maurizi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Maria Grazia Ammendolia
- National Center for Innovative Technologies in Public Health, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Catia Longhi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (G.B.); (L.M.); (C.L.); (B.A.)
| |
Collapse
|
14
|
Chang YC, Liu WL, Fang PH, Li JC, Liu KL, Huang JL, Chen HW, Kao CF, Chen CH. Effect of C-type lectin 16 on dengue virus infection in Aedes aegypti salivary glands. PNAS NEXUS 2024; 3:pgae188. [PMID: 38813522 PMCID: PMC11134184 DOI: 10.1093/pnasnexus/pgae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/29/2024] [Indexed: 05/31/2024]
Abstract
C-type lectins (CTLs) are a family of carbohydrate-binding proteins and an important component of mosquito saliva. Although CTLs play key roles in immune activation and viral pathogenesis, little is known about their role in regulating dengue virus (DENV) infection and transmission. In this study, we established a homozygous CTL16 knockout Aedes aegypti mutant line using CRISPR/Cas9 to study the interaction between CTL16 and viruses in mosquito vectors. Furthermore, mouse experiments were conducted to confirm the transmission of DENV by CTL16-/- A. aegypti mutants. We found that CTL16 was mainly expressed in the medial lobe of the salivary glands (SGs) in female A. aegypti. CTL16 knockout increased DENV replication and accumulation in the SGs of female A. aegypti, suggesting that CTL16 plays an important role in DENV transmission. We also found a reduced expression of immunodeficiency and Janus kinase/signal transducer and activator of transcription pathway components correlated with increased DENV viral titer, infection rate, and transmission efficiency in the CTL16 mutant strain. The findings of this study provide insights not only for guiding future investigations on the influence of CTLs on immune responses in mosquitoes but also for developing novel mutants that can be used as vector control tools.
Collapse
Affiliation(s)
- Ya-Chen Chang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Pai-Hsiang Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Kun-Lin Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jau-Ling Huang
- Department of Bioscience Technology, Chang Jung Christian University, Tainan 711301, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Chun-Hong Chen
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| |
Collapse
|
15
|
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
|
16
|
Lopez-Perez M, Jain A, Davies DH, Vásquez-Jiménez JM, Herrera SM, Oñate J, Felgner PL, Herrera S, Arévalo-Herrera M. Profiling the antibody response of humans protected by immunization with Plasmodium vivax radiation-attenuated sporozoites. Sci Rep 2024; 14:2790. [PMID: 38307966 PMCID: PMC10837454 DOI: 10.1038/s41598-024-53175-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 02/04/2024] Open
Abstract
Malaria sterile immunity has been reproducibly induced by immunization with Plasmodium radiation-attenuated sporozoites (RAS). Analyses of sera from RAS-immunized individuals allowed the identification of P. falciparum antigens, such as the circumsporozoite protein (CSP), the basis for the RTS, S and R21Matrix-M vaccines. Similar advances in P. vivax (Pv) vaccination have been elusive. We previously reported 42% (5/12) of sterile protection in malaria-unexposed, Duffy-positive (Fy +) volunteers immunized with PvRAS followed by a controlled human malaria infection (CHMI). Using a custom protein microarray displaying 515 Pv antigens, we found a significantly higher reactivity to PvCSP and one hypothetical protein (PVX_089630) in volunteers protected against P. vivax infection. In mock-vaccinated Fy + volunteers, a strong antibody response to CHMI was also observed. Although the Fy- volunteers immunized with non-irradiated Pv-infected mosquitoes (live sporozoites) did not develop malaria after CHMI, they recognized a high number of antigens, indicating the temporary presence of asexual parasites in peripheral blood. Together, our findings contribute to the understanding of the antibody response to P. vivax infection and allow the identification of novel parasite antigens as vaccine candidates.Trial registration: ClinicalTrials.gov number: NCT01082341.
Collapse
Affiliation(s)
- Mary Lopez-Perez
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
| | - Aarti Jain
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - D Huw Davies
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | | | | | | | - Philip L Felgner
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - Sócrates Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
- Caucaseco Scientific Research Center, Cali, Colombia
| | - Myriam Arévalo-Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia.
- Caucaseco Scientific Research Center, Cali, Colombia.
| |
Collapse
|
17
|
Shrivastava G, Valenzuela-Leon PC, Botello K, Calvo E. Aedes aegypti saliva modulates inflammasome activation and facilitates flavivirus infection in vitro. iScience 2024; 27:108620. [PMID: 38188518 PMCID: PMC10770497 DOI: 10.1016/j.isci.2023.108620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/16/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Mosquito borne flaviviruses such as dengue and Zika represent a major public health problem due to globalization and propagation of susceptible vectors worldwide. Vertebrate host responses to dengue and Zika infections include the processing and release of pro-inflammatory cytokines through the activation of inflammasomes, resulting in disease severity and fatality. Mosquito saliva can facilitate pathogen infection by downregulating the host's immune response. However, the role of mosquito saliva in modulating host innate immune responses remains largely unknown. Here, we show that mosquito salivary gland extract (SGE) inhibits dengue and Zika virus-induced inflammasome activation by reducing NLRP3 expression, Caspase-1 activation, and 1L-1β secretion in cultured human and mice macrophages. As a result, we observe that SGE inhibits virus detection in the early phase of infection. This study provides important insights into how mosquito saliva modulates host innate immunity during viral infection.
Collapse
Affiliation(s)
- Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Karina Botello
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway Room 2W09, Bethesda, MD, USA
| |
Collapse
|
18
|
Howell MM, Olajiga OM, Cardenas JC, Parada-Higuera CA, Gonzales-Pabon MU, Gutierrez-Silva LY, Jaimes-Villamizar L, Werner BM, Shaffer JG, Manuzak JA, Londono-Renteria B. Mosquito Salivary Antigens and Their Relationship to Dengue and P. vivax Malaria. Pathogens 2024; 13:52. [PMID: 38251359 PMCID: PMC10818852 DOI: 10.3390/pathogens13010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/10/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
In tropical areas, the simultaneous transmission of multiple vector-borne diseases is common due to ecological factors shared by arthropod vectors. Malaria and dengue virus, transmitted by Anopheles and Aedes mosquitoes, respectively, are among the top vector-borne diseases that cause significant morbidity and mortality in endemic areas. Notably, tropical areas often have suitable conditions for the co-existence of these mosquito species, highlighting the importance of identifying markers that accurately indicate the risk of acquiring each specific disease entity. Aedes are daytime-biting mosquitoes, while Anopheles preferentially bite during the night. These biting patterns raise the possibility of concurrent exposure to bites from both species. This is important because mosquito saliva, deposited in the skin during blood feeding, induces immune responses that modulate pathogen establishment and infection. Previous studies have focused on characterizing such effects on the vector-pathogen interface for an individual pathogen and its mosquito vector. In this study, we evaluated associations between immune responses to salivary proteins from non-dengue and non-malaria vector mosquito species with clinical characteristics of malaria and dengue, respectively. Surprisingly, antibody responses against Anopheles antigens in dengue patients correlated with red blood cell count and hematocrit, while antibody responses against Aedes proteins were associated with platelet count in malaria patients. Our data indicate that concurrent exposure to multiple disease-carrying mosquito vectors and their salivary proteins with differing immunomodulatory properties could influence the transmission, pathogenesis, and clinical presentation of malaria, dengue fever, and other vector-borne illnesses.
Collapse
Affiliation(s)
- McKenna M. Howell
- Arbovirology Laboratory, Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA; (M.M.H.); (J.C.C.)
| | - Olayinka M. Olajiga
- Arbovirology Laboratory, Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA; (M.M.H.); (J.C.C.)
| | - Jenny C. Cardenas
- Arbovirology Laboratory, Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA; (M.M.H.); (J.C.C.)
| | | | | | | | | | - Brett M. Werner
- College of Science and Technology, Bellevue University, Bellevue, NE 68005, USA;
| | - Jeffrey G. Shaffer
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA;
| | - Jennifer A. Manuzak
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Berlin Londono-Renteria
- Arbovirology Laboratory, Department of Tropical Medicine and Infectious Disease, Tulane University, New Orleans, LA 70112, USA; (M.M.H.); (J.C.C.)
| |
Collapse
|
19
|
Cantillo JF, Garcia E, Fernandez-Caldas E, Puerta L. Mosquito allergy: Immunological aspects and clinical management. Mol Immunol 2023; 164:153-158. [PMID: 38039596 DOI: 10.1016/j.molimm.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Mosquito allergy has been conceived as the cutaneous reactions that appears during and after mosquito biting process; a perception that is supported by several scientific research. Additional data have led to conceive that other manifestations of allergic responses may occur as a cause of the exposure to somatic mosquito allergens. Two main phenotypes of mosquito allergy are identifiable: the cutaneous allergic reactions, induced by salivary allergens, and other manifestations of the allergic responses such as asthma and allergic rhino conjunctivitis that are caused by somatic allergens. The cutaneous reactions have kept the focus of attention of the scientific community. It appears as skin lesions that resembles the phenotype of papular urticaria with a defined natural history of the disease. Although these two phenotypes of mosquito allergy seem to be well differentiated in terms of the allergens that are involved and the routes of exposures, other factors such as geographical distribution, may participate. Mosquitoes have adapted to the host immune response against bites, producing immunomodulatory molecules that counteract such defensive response. The role that the immunomodulatory molecules have on the allergic immune response has not been studied yet and it is still not known if affects all mosquito allergy phenotypes. Only a few studies of allergen specific immunotherapy for cutaneous allergic reactions induced by mosquito bites have been done, and none for respiratory allergic responses. The clinical practice focuses on symptom management and avoiding mosquito bites as much as possible. Avoiding mosquitoes, using different well described methods, is still the best option to limit contact with these insects. The lack of knowledge of mosquito allergy have raised several questions that affects the clinical management of this allergic disease, from its diagnosis, prevention and immunotherapy.
Collapse
Affiliation(s)
| | - Elizabeth Garcia
- Faculty of Medicine, Universidad de los Andes, Allergy Section, Fundación Santa Fe de Bogotá, UNIMEQ ORL, Bogotá, Colombia
| | | | - Leonardo Puerta
- Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| |
Collapse
|
20
|
Alexander LT, Durairaj J, Kryshtafovych A, Abriata LA, Bayo Y, Bhabha G, Breyton C, Caulton SG, Chen J, Degroux S, Ekiert DC, Erlandsen BS, Freddolino PL, Gilzer D, Greening C, Grimes JM, Grinter R, Gurusaran M, Hartmann MD, Hitchman CJ, Keown JR, Kropp A, Kursula P, Lovering AL, Lemaitre B, Lia A, Liu S, Logotheti M, Lu S, Markússon S, Miller MD, Minasov G, Niemann HH, Opazo F, Phillips GN, Davies OR, Rommelaere S, Rosas‐Lemus M, Roversi P, Satchell K, Smith N, Wilson MA, Wu K, Xia X, Xiao H, Zhang W, Zhou ZH, Fidelis K, Topf M, Moult J, Schwede T. Protein target highlights in CASP15: Analysis of models by structure providers. Proteins 2023; 91:1571-1599. [PMID: 37493353 PMCID: PMC10792529 DOI: 10.1002/prot.26545] [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: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 07/27/2023]
Abstract
We present an in-depth analysis of selected CASP15 targets, focusing on their biological and functional significance. The authors of the structures identify and discuss key protein features and evaluate how effectively these aspects were captured in the submitted predictions. While the overall ability to predict three-dimensional protein structures continues to impress, reproducing uncommon features not previously observed in experimental structures is still a challenge. Furthermore, instances with conformational flexibility and large multimeric complexes highlight the need for novel scoring strategies to better emphasize biologically relevant structural regions. Looking ahead, closer integration of computational and experimental techniques will play a key role in determining the next challenges to be unraveled in the field of structural molecular biology.
Collapse
Affiliation(s)
- Leila T. Alexander
- BiozentrumUniversity of BaselBaselSwitzerland
- Computational Structural BiologySIB Swiss Institute of BioinformaticsBaselSwitzerland
| | - Janani Durairaj
- BiozentrumUniversity of BaselBaselSwitzerland
- Computational Structural BiologySIB Swiss Institute of BioinformaticsBaselSwitzerland
| | | | - Luciano A. Abriata
- School of Life SciencesÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Yusupha Bayo
- Department of BiosciencesUniversity of MilanoMilanItaly
- IBBA‐CNR Unit of MilanoInstitute of Agricultural Biology and BiotechnologyMilanItaly
| | - Gira Bhabha
- Department of Cell BiologyNew York University School of MedicineNew YorkNew YorkUSA
| | | | | | - James Chen
- Department of Cell BiologyNew York University School of MedicineNew YorkNew YorkUSA
| | | | - Damian C. Ekiert
- Department of Cell BiologyNew York University School of MedicineNew YorkNew YorkUSA
- Department of MicrobiologyNew York University School of MedicineNew YorkNew YorkUSA
| | - Benedikte S. Erlandsen
- Wellcome Centre for Cell BiologyInstitute of Cell Biology, University of EdinburghEdinburghUK
| | - Peter L. Freddolino
- Department of Biological Chemistry, Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
| | - Dominic Gilzer
- Department of ChemistryBielefeld UniversityBielefeldGermany
| | - Chris Greening
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Securing Antarctica's Environmental FutureMonash UniversityClaytonVictoriaAustralia
- Centre to Impact AMRMonash UniversityClaytonVictoriaAustralia
- ARC Research Hub for Carbon Utilisation and RecyclingMonash UniversityClaytonVictoriaAustralia
| | - Jonathan M. Grimes
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Rhys Grinter
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
- Centre for Electron Microscopy of Membrane ProteinsMonash Institute of Pharmaceutical SciencesParkvilleVictoriaAustralia
| | - Manickam Gurusaran
- Wellcome Centre for Cell BiologyInstitute of Cell Biology, University of EdinburghEdinburghUK
| | - Marcus D. Hartmann
- Max Planck Institute for BiologyTübingenGermany
- Interfaculty Institute of Biochemistry, University of TübingenTübingenGermany
| | - Charlie J. Hitchman
- Department of Molecular and Cell Biology, Leicester Institute of Structural and Chemical BiologyUniversity of LeicesterLeicesterUK
| | - Jeremy R. Keown
- Division of Structural Biology, Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Ashleigh Kropp
- Department of Microbiology, Biomedicine Discovery InstituteMonash UniversityClaytonVictoriaAustralia
| | - Petri Kursula
- Department of BiomedicineUniversity of BergenBergenNorway
- Faculty of Biochemistry and Molecular Medicine & Biocenter OuluUniversity of OuluOuluFinland
| | | | - Bruno Lemaitre
- School of Life SciencesÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Andrea Lia
- Department of Molecular and Cell Biology, Leicester Institute of Structural and Chemical BiologyUniversity of LeicesterLeicesterUK
- ISPA‐CNR Unit of LecceInstitute of Sciences of Food ProductionLecceItaly
| | - Shiheng Liu
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Maria Logotheti
- Max Planck Institute for BiologyTübingenGermany
- Interfaculty Institute of Biochemistry, University of TübingenTübingenGermany
- Present address:
Institute of BiochemistryUniversity of GreifswaldGreifswaldGermany
| | - Shuze Lu
- Lanzhou University School of Life SciencesLanzhouChina
| | | | | | - George Minasov
- Department of Microbiology‐ImmunologyNorthwestern Feinberg School of MedicineChicagoIllinoisUSA
| | | | - Felipe Opazo
- NanoTag Biotechnologies GmbHGöttingenGermany
- Institute of Neuro‐ and Sensory PhysiologyUniversity of Göttingen Medical CenterGöttingenGermany
- Center for Biostructural Imaging of Neurodegeneration (BIN)University of Göttingen Medical CenterGöttingenGermany
| | - George N. Phillips
- Department of BiosciencesRice UniversityHoustonTexasUSA
- Department of ChemistryRice UniversityHoustonTexasUSA
| | - Owen R. Davies
- Wellcome Centre for Cell BiologyInstitute of Cell Biology, University of EdinburghEdinburghUK
| | - Samuel Rommelaere
- School of Life SciencesÉcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Monica Rosas‐Lemus
- Department of Microbiology‐ImmunologyNorthwestern Feinberg School of MedicineChicagoIllinoisUSA
- Present address:
Department of Molecular Genetics and MicrobiologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Pietro Roversi
- IBBA‐CNR Unit of MilanoInstitute of Agricultural Biology and BiotechnologyMilanItaly
- Department of Molecular and Cell Biology, Leicester Institute of Structural and Chemical BiologyUniversity of LeicesterLeicesterUK
| | - Karla Satchell
- Department of Microbiology‐ImmunologyNorthwestern Feinberg School of MedicineChicagoIllinoisUSA
| | - Nathan Smith
- Department of Biochemistry and the Redox Biology CenterUniversity of NebraskaLincolnNebraskaUSA
| | - Mark A. Wilson
- Department of Biochemistry and the Redox Biology CenterUniversity of NebraskaLincolnNebraskaUSA
| | - Kuan‐Lin Wu
- Department of ChemistryRice UniversityHoustonTexasUSA
| | - Xian Xia
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Han Xiao
- Department of BiosciencesRice UniversityHoustonTexasUSA
- Department of ChemistryRice UniversityHoustonTexasUSA
- Department of BioengineeringRice UniversityHoustonTexasUSA
| | - Wenhua Zhang
- Lanzhou University School of Life SciencesLanzhouChina
| | - Z. Hong Zhou
- Department of Microbiology, Immunology, and Molecular GeneticsUniversity of CaliforniaLos AngelesCaliforniaUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCaliforniaUSA
| | | | - Maya Topf
- University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
- Centre for Structural Systems BiologyLeibniz‐Institut für Virologie (LIV)HamburgGermany
| | - John Moult
- Department of Cell Biology and Molecular Genetics, Institute for Bioscience and Biotechnology ResearchUniversity of MarylandRockvilleMarylandUSA
| | - Torsten Schwede
- BiozentrumUniversity of BaselBaselSwitzerland
- Computational Structural BiologySIB Swiss Institute of BioinformaticsBaselSwitzerland
| |
Collapse
|
21
|
Henriques P, Rosa A, Caldeira-Araújo H, Soares P, Vigário AM. Flying under the radar - impact and factors influencing asymptomatic DENV infections. Front Cell Infect Microbiol 2023; 13:1284651. [PMID: 38076464 PMCID: PMC10704250 DOI: 10.3389/fcimb.2023.1284651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
The clinical outcome of DENV and other Flaviviruses infections represents a spectrum of severity that ranges from mild manifestations to severe disease, which can ultimately lead to death. Nonetheless, most of these infections result in an asymptomatic outcome that may play an important role in the persistent circulation of these viruses. Also, although little is known about the mechanisms that lead to these asymptomatic infections, they are likely the result of a complex interplay between viral and host factors. Specific characteristics of the infecting viral strain, such as its replicating efficiency, coupled with host factors, like gene expression of key molecules involved in the immune response or in the protection against disease, are among crucial factors to study. This review revisits recent data on factors that may contribute to the asymptomatic outcome of the world's widespread DENV, highlighting the importance of silent infections in the transmission of this pathogen and the immune status of the host.
Collapse
Affiliation(s)
- Paulo Henriques
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Alexandra Rosa
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Pedro Soares
- Department of Biology, CBMA (Centre of Molecular and Environmental Biology), Braga, Portugal
- Department of Biology, Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, Braga, Portugal
| | - Ana Margarida Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
22
|
Watson FN, Shears MJ, Kalata AC, Duncombe CJ, Seilie AM, Chavtur C, Conrad E, Talavera IC, Raappana A, Sather DN, Chakravarty S, Sim BKL, Hoffman SL, Tsuji M, Murphy SC. Ultra-low volume intradermal administration of radiation-attenuated sporozoites with the glycolipid adjuvant 7DW8-5 completely protects mice against malaria. RESEARCH SQUARE 2023:rs.3.rs-3243319. [PMID: 37609210 PMCID: PMC10441511 DOI: 10.21203/rs.3.rs-3243319/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Malaria is caused by Plasmodium parasites and was responsible for over 247 million infections and 619,000 deaths in 2021. Radiation-attenuated sporozoite (RAS) vaccines can completely prevent blood stage infection by inducing protective liver-resident memory CD8+ T cells. Such T cells can be induced by 'prime-and-trap' vaccination, which here combines DNA priming against the P. yoelii circumsporozoite protein (CSP) with a subsequent intravenous (IV) dose of liver-homing RAS to "trap" the activated and expanding T cells in the liver. Prime-and-trap confers durable protection in mice, and efforts are underway to translate this vaccine strategy to the clinic. However, it is unclear whether the RAS trapping dose must be strictly administered by the IV route. Here we show that intradermal (ID) RAS administration can be as effective as IV administration if RAS are co-administrated with the glycolipid adjuvant 7DW8-5 in an ultra-low inoculation volume. In mice, the co-administration of RAS and 7DW8-5 in ultra-low ID volumes (2.5 μL) was completely protective and dose sparing compared to standard volumes (10-50 μL) and induced protective levels of CSP-specific CD8+ T cells in the liver. Our finding that adjuvants and ultra-low volumes are required for ID RAS efficacy may explain why prior reports about higher volumes of unadjuvanted ID RAS proved less effective. The ID route may offer significant translational advantages over the IV route and could improve sporozoite vaccine development.
Collapse
|
23
|
Crawford JM, Buechlein AM, Moline DA, Rusch DB, Hardy RW. Host Derivation of Sindbis Virus Influences Mammalian Type I Interferon Response to Infection. Viruses 2023; 15:1685. [PMID: 37632027 PMCID: PMC10458878 DOI: 10.3390/v15081685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Arboviruses are defined by their ability to replicate in both mosquito vectors and mammalian hosts. There is good evidence that arboviruses "prime" their progeny for infection of the next host, such as via differential glycosylation of their outer glycoproteins or packaging of host ribosomal subunits. We and others have previously shown that mosquito-derived viruses more efficiently infect mammalian cells than mammalian-derived viruses. These observations are consistent with arboviruses acquiring host-specific adaptations, and we hypothesized that a virus derived from either the mammalian host or mosquito vector elicits different responses when infecting the mammalian host. Here, we perform an RNA-sequencing analysis of the transcriptional response of Human Embryonic Kidney 293 (HEK-293) cells to infection with either mosquito (Aedes albopictus, C7/10)- or mammalian (Baby Hamster Kidney, BHK-21)-derived Sindbis virus (SINV). We show that the C7/10-derived virus infection leads to a more robust transcriptional response in HEK-293s compared to infection with the BHK-derived virus. Surprisingly, despite more efficient infection, we found an increase in interferon-β (IFN-β) and interferon-stimulated gene (ISG) transcripts in response to the C7/10-derived virus infection versus the BHK-derived virus infection. However, translation of interferon-stimulated genes was lower in HEK-293s infected with the C7/10-derived virus, starkly contrasting with the transcriptional response. This inhibition of ISG translation is reflective of a more rapid overall shut-off of host cell translation following infection with the C7/10-derived virus. Finally, we show that the C7/10-derived virus infection of HEK-293 cells leads to elevated levels of phosphorylated eukaryotic translation elongation factor-2 (eEF2), identifying a potential mechanism leading to the more rapid shut-off of host translation. We postulate that the rapid shut-off of host translation in mammalian cells infected with the mosquito-derived virus acts to counter the IFN-β-stimulated transcriptional response.
Collapse
Affiliation(s)
- John M. Crawford
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; (J.M.C.); (D.A.M.)
| | - Aaron M. Buechlein
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA; (A.M.B.); (D.B.R.)
| | - Davis A. Moline
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; (J.M.C.); (D.A.M.)
| | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA; (A.M.B.); (D.B.R.)
| | - Richard W. Hardy
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; (J.M.C.); (D.A.M.)
| |
Collapse
|
24
|
Seavey CE, Doshi M, Panarello AP, Felice MA, Dickerson AK, Jewett MW, Willenberg BJ. Engineered Human Tissue as A New Platform for Mosquito Bite-Site Biology Investigations. INSECTS 2023; 14:514. [PMID: 37367330 PMCID: PMC10299109 DOI: 10.3390/insects14060514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023]
Abstract
Vector-borne diseases transmitted through the bites of hematophagous arthropods, such as mosquitoes, continue to be a significant threat to human health globally. Transmission of disease by biting arthropod vectors includes interactions between (1) saliva expectorated by a vector during blood meal acquisition from a human host, (2) the transmitted vector-borne pathogens, and (3) host cells present at the skin bite site. Currently, the investigation of bite-site biology is challenged by the lack of model 3D human skin tissues for in vitro analyses. To help fill this gap, we have used a tissue engineering approach to develop new stylized human dermal microvascular bed tissue approximates-complete with warm blood-built with 3D capillary alginate gel (Capgel) biomaterial scaffolds. These engineered tissues, termed a Biologic Interfacial Tissue-Engineered System (BITES), were cellularized with either human dermal fibroblasts (HDFs) or human umbilical vein endothelial cells (HUVECs). Both cell types formed tubular microvessel-like tissue structures of oriented cells (82% and 54% for HDFs and HUVECs, respectively) lining the unique Capgel parallel capillary microstructures. Female Aedes (Ae.) aegypti mosquitoes, a prototypic hematophagous biting vector arthropod, swarmed, bit, and probed blood-loaded HDF BITES microvessel bed tissues that were warmed (34-37 °C), acquiring blood meals in 151 ± 46 s on average, with some ingesting ≳4 µL or more of blood. Further, these tissue-engineered constructs could be cultured for at least three (3) days following blood meal acquisitions. Altogether, these studies serve as a powerful proof-of-concept demonstration of the innovative BITES platform and indicate its potential for the future investigation of arthropod bite-site cellular and molecular biology.
Collapse
Affiliation(s)
- Corey E. Seavey
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Mona Doshi
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Andrew P. Panarello
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Michael A. Felice
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Andrew K. Dickerson
- Department of Mechanical, Aerospace, and Biomedical Engineering, Tickle College of Engineering, University of Tennessee, Knoxville, TN 37996, USA
| | - Mollie W. Jewett
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| | - Bradley J. Willenberg
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, USA
| |
Collapse
|
25
|
Marín-López A, Raduwan H, Chen TY, Utrilla-Trigo S, Wolfhard DP, Fikrig E. Mosquito Salivary Proteins and Arbovirus Infection: From Viral Enhancers to Potential Targets for Vaccines. Pathogens 2023; 12:371. [PMID: 36986293 PMCID: PMC10054260 DOI: 10.3390/pathogens12030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/13/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Arthropod-borne viruses present important public health challenges worldwide. Viruses such as DENV, ZIKV, and WNV are of current concern due to an increasing incidence and an expanding geographic range, generating explosive outbreaks even in non-endemic areas. The clinical signs associated with infection from these arboviruses are often inapparent, mild, or nonspecific, but occasionally develop into serious complications marked by rapid onset, tremors, paralysis, hemorrhagic fever, neurological alterations, or death. They are predominately transmitted to humans through mosquito bite, during which saliva is inoculated into the skin to facilitate blood feeding. A new approach to prevent arboviral diseases has been proposed by the observation that arthropod saliva facilitates transmission of pathogens. Viruses released within mosquito saliva may more easily initiate host invasion by taking advantage of the host's innate and adaptive immune responses to saliva. This provides a rationale for creating vaccines against mosquito salivary proteins, especially because of the lack of licensed vaccines against most of these viruses. This review aims to provide an overview of the effects on the host immune response by the mosquito salivary proteins and how these phenomena alter the infection outcome for different arboviruses, recent attempts to generate mosquito salivary-based vaccines against flavivirus including DENV, ZIKV, and WNV, and the potential benefits and pitfalls that this strategy involves.
Collapse
Affiliation(s)
- Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Sergio Utrilla-Trigo
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
- Center for Animal Health Research (CISA-INIA/CSIC), 28130 Madrid, Spain
| | - David P. Wolfhard
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
- Faculty of Engineering Sciences, Institute of Pharmacy and Molecular Biotechnology, 69120 Heidelberg, Germany
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
26
|
Native structure of mosquito salivary protein uncovers domains relevant to pathogen transmission. Nat Commun 2023; 14:899. [PMID: 36797290 PMCID: PMC9935623 DOI: 10.1038/s41467-023-36577-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Female mosquitoes inject saliva into vertebrate hosts during blood feeding. This process transmits mosquito-borne human pathogens that collectively cause ~1,000,000 deaths/year. Among the most abundant and conserved proteins secreted by female salivary glands is a high-molecular weight protein called salivary gland surface protein 1 (SGS1) that facilitates pathogen transmission, but its mechanism remains elusive. Here, we determine the native structure of SGS1 by the cryoID approach, showing that the 3364 amino-acid protein has a Tc toxin-like Rhs/YD shell, four receptor domains, and a set of C-terminal daisy-chained helices. These helices are partially shielded inside the Rhs/YD shell and poised to transform into predicted transmembrane helices. This transformation, and the numerous receptor domains on the surface of SGS1, are likely key in facilitating sporozoite/arbovirus invasion into the salivary glands and manipulating the host's immune response.
Collapse
|
27
|
Spencer Clinton JL, Vogt MB, Kneubehl AR, Hibl BM, Paust S, Rico-Hesse R. Sialokinin in mosquito saliva shifts human immune responses towards intracellular pathogens. PLoS Negl Trop Dis 2023; 17:e0011095. [PMID: 36735632 PMCID: PMC9897557 DOI: 10.1371/journal.pntd.0011095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
Mosquito saliva is a mix of numerous proteins that are injected into the skin while the mosquito searches for a blood meal. While mosquito saliva is known to be immunogenic, the salivary components driving these immune responses, as well as the types of immune responses that occur, are not well characterized. We investigated the effects of one potential immunomodulatory mosquito saliva protein, sialokinin, on the human immune response. We used flow cytometry to compare human immune cell populations between humanized mice bitten by sialokinin knockout mosquitoes or injected with sialokinin, and compared them to those bitten by wild-type mosquitoes, unbitten, or saline-injected control mice. Humanized mice received 4 mosquito bites or a single injection, were euthanized after 7 days, and skin, spleen, bone marrow, and blood were harvested for immune cell profiling. Our results show that bites from sialokinin knockout mosquitoes induced monocyte and macrophage populations in the skin, blood, bone marrow, and spleens, and primarily affected CD11c- cell populations. Other increased immune cells included plasmacytoid dendritic cells in the blood, natural killer cells in the skin and blood, and CD4+ T cells in all samples analyzed. Conversely, we observed that mice bitten with sialokinin knockout mosquitoes had decreased NKT cell populations in the skin, and fewer B cells in the blood, spleen, and bone marrow. Taken together, we demonstrated that sialokinin knockout saliva induces elements of a TH1 cellular immune response, suggesting that the sialokinin peptide is inducing a TH2 cellular immune response during wild-type mosquito biting. These findings are an important step towards understanding how mosquito saliva modulates the human immune system and which components of saliva may be critical for arboviral infection. By identifying immunomodulatory salivary proteins, such as sialokinin, we can develop vaccines against mosquito saliva components and direct efforts towards blocking arboviral infections.
Collapse
Affiliation(s)
- Jennifer L. Spencer Clinton
- 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
| | - Alexander R. Kneubehl
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Brianne M. Hibl
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Silke Paust
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California, United States of America
| | - Rebecca Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
28
|
Guerrero D, Vo HTM, Lon C, Bohl JA, Nhik S, Chea S, Man S, Sreng S, Pacheco AR, Ly S, Sath R, Lay S, Missé D, Huy R, Leang R, Kry H, Valenzuela JG, Oliveira F, Cantaert T, Manning JE. Evaluation of cutaneous immune response in a controlled human in vivo model of mosquito bites. Nat Commun 2022; 13:7036. [PMID: 36396947 PMCID: PMC9672097 DOI: 10.1038/s41467-022-34534-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne viruses are a growing global threat. Initial viral inoculation occurs in the skin via the mosquito 'bite', eliciting immune responses that shape the establishment of infection and pathogenesis. Here we assess the cutaneous innate and adaptive immune responses to controlled Aedes aegypti feedings in humans living in Aedes-endemic areas. In this single-arm, cross-sectional interventional study (trial registration #NCT04350905), we enroll 30 healthy adult participants aged 18 to 45 years of age from Cambodia between October 2020 and January 2021. We perform 3-mm skin biopsies at baseline as well as 30 min, 4 h, and 48 h after a controlled feeding by uninfected Aedes aegypti mosquitos. The primary endpoints are measurement of changes in early and late innate responses in bitten vs unbitten skin by gene expression profiling, immunophenotyping, and cytokine profiling. The results reveal induction of neutrophil degranulation and recruitment of skin-resident dendritic cells and M2 macrophages. As the immune reaction progresses T cell priming and regulatory pathways are upregulated along with a shift to Th2-driven responses and CD8+ T cell activation. Stimulation of participants' bitten skin cells with Aedes aegypti salivary gland extract results in reduced pro-inflammatory cytokine production. These results identify key immune genes, cell types, and pathways in the human response to mosquito bites and can be leveraged to inform and develop novel therapeutics and vector-targeted vaccine candidates to interfere with vector-mediated disease.
Collapse
Affiliation(s)
- David Guerrero
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Chanthap Lon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Jennifer A Bohl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sreynik Nhik
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sophana Chea
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Somnang Man
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Andrea R Pacheco
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sokna Ly
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Rathanak Sath
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sokchea Lay
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34000, Montpellier, France
| | - Rekol Huy
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Hok Kry
- Kampong Speu Provincial District, Ministry of Health, Phnom Penh, Cambodia
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tineke Cantaert
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Jessica E Manning
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
29
|
Olajiga OM, Marin-Lopez A, Cardenas JC, Gutierrez-Silva LY, Gonzales-Pabon MU, Maldonado-Ruiz LP, Worges M, Fikrig E, Park Y, Londono-Renteria B. Aedes aegypti anti-salivary proteins IgG levels in a cohort of DENV-like symptoms subjects from a dengue-endemic region in Colombia. FRONTIERS IN EPIDEMIOLOGY 2022; 2:1002857. [PMID: 38455331 PMCID: PMC10910902 DOI: 10.3389/fepid.2022.1002857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 03/09/2024]
Abstract
Dengue fever, caused by the dengue virus (DENV), is currently a threat to about half of the world's population. DENV is mainly transmitted to the vertebrate host through the bite of a female Aedes mosquito while taking a blood meal. During this process, salivary proteins are introduced into the host skin and blood to facilitate blood acquisition. These salivary proteins modulate both local (skin) and systemic immune responses. Several salivary proteins have been identified as immunogenic inducing the production of antibodies with some of those proteins also displaying immunomodulatory properties enhancing arboviral infections. IgG antibody responses against salivary gland extracts of a diverse number of mosquitoes, as well as antibody responses against the Ae. aegypti peptide, Nterm-34 kDa, have been suggested as biomarkers of human exposure to mosquito bites while antibodies against AgBR1 and NeSt1 proteins have been investigated for their potential protective effect against Zika virus (ZIKV) and West Nile virus infections. Thus, we were interested in evaluating whether IgG antibodies against AgBR1, NeSt1, Nterm-34 kDa peptide, and SGE were associated with DENV infections and clinical characteristics. For this, we tested samples from volunteers living in a dengue fever endemic area in Colombia in 2019 for the presence of IgG antibodies against those salivary proteins and peptides using an ELISA test. Results from this pilot study suggest an involvement of antibody responses against salivary proteins in dengue disease progression.
Collapse
Affiliation(s)
- Olayinka M. Olajiga
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Alejandro Marin-Lopez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jenny C. Cardenas
- Laboratorio Clínico, Hospital Local Los Patios, Los Patios, Colombia
| | | | | | | | - Matt Worges
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University of New Orleans, New Orleans, LA, United States
| | - Erol Fikrig
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Yoonseong Park
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Berlin Londono-Renteria
- Department of Entomology, Kansas State University, Manhattan, KS, United States
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University of New Orleans, New Orleans, LA, United States
| |
Collapse
|
30
|
Fonseka CL, Hardman CS, Woo J, Singh R, Nahler J, Yang J, Chen YL, Kamaladasa A, Silva T, Salimi M, Gray N, Dong T, Malavige GN, Ogg GS. Dengue virus co-opts innate type 2 pathways to escape early control of viral replication. Commun Biol 2022; 5:735. [PMID: 35869167 PMCID: PMC9306424 DOI: 10.1038/s42003-022-03682-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 07/06/2022] [Indexed: 12/13/2022] Open
Abstract
Mast cell products and high levels of type 2 cytokines are associated with severe dengue disease. Group 2 innate lymphoid cells (ILC2) are type-2 cytokine-producing cells that are activated by epithelial cytokines and mast cell-derived lipid mediators. Through ex vivo RNAseq analysis, we observed that ILC2 are activated during acute dengue viral infection, and show an impaired type I-IFN signature in severe disease. We observed that circulating ILC2 are permissive for dengue virus infection in vivo and in vitro, particularly when activated through prostaglandin D2 (PGD2). ILC2 underwent productive dengue virus infection, which was inhibited through CRTH2 antagonism. Furthermore, exogenous IFN-β induced expression of type I-IFN responsive anti-viral genes by ILC2. PGD2 downregulated type I-IFN responsive gene and protein expression; and urinary prostaglandin D2 metabolite levels were elevated in severe dengue. Moreover, supernatants from activated ILC2 enhanced monocyte infection in a GM-CSF and mannan-dependent manner. Our results indicate that dengue virus co-opts an innate type 2 environment to escape early type I-IFN control and facilitate viral dissemination. PGD2 downregulates type I-IFN induced anti-viral responses in ILC2. CRTH2 antagonism may be a therapeutic strategy for dengue-associated disease.
Collapse
Affiliation(s)
- Chathuranga L Fonseka
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Medicine, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | - Clare S Hardman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jeongmin Woo
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Randeep Singh
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Janina Nahler
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jiahe Yang
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Yi-Ling Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Achala Kamaladasa
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Tehani Silva
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- General Sir John Kotelawala Defence University, Rathmalana, Sri Lanka
| | - Maryam Salimi
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicki Gray
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Tao Dong
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Gathsaurie N Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Graham S Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK.
| |
Collapse
|
31
|
Shrivastava G, Valenzuela-Leon PC, Chagas AC, Kern O, Botello K, Zhang Y, Martin-Martin I, Oliveira MB, Tirloni L, Calvo E. Alboserpin, the Main Salivary Anticoagulant from the Disease Vector Aedes albopictus, Displays Anti-FXa-PAR Signaling In Vitro and In Vivo. Immunohorizons 2022; 6:373-383. [PMID: 35738824 PMCID: PMC10753553 DOI: 10.4049/immunohorizons.2200045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/19/2022] Open
Abstract
Blood-feeding arthropods secrete potent salivary molecules, which include platelet aggregation inhibitors, vasodilators, and anticoagulants. Among these molecules, Alboserpin, the major salivary anticoagulant from the mosquito vector Aedes albopictus, is a specific inhibitor of the human coagulation factor Xa (FXa). In this study, we investigated the anti-inflammatory properties of Alboserpin, in vitro and in vivo. In vitro, Alboserpin inhibited FXa-induced protease-activated receptor (PAR)-1, PAR-2, PAR-3, VCAM, ICAM, and NF-κB gene expression in primary dermal microvascular endothelial cells. Alboserpin also prevented FXa-stimulated ERK1/2 gene expression and subsequent inflammatory cytokine release (MCP-1, TNF-α, IL-6, IL-8, IL-1β, IL-18). In vivo, Alboserpin reduced paw edema induced by FXa and subsequent release of inflammatory cytokines (CCL2, MCP-1, IL-1α, IL-6, IL-1β). Alboserpin also reduced FXa-induced endothelial permeability in vitro and in vivo. These findings show that Alboserpin is a potent anti-inflammatory molecule, in vivo and in vitro, and may play a significant role in blood feeding.
Collapse
Affiliation(s)
- Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Karina Botello
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Yixiang Zhang
- Protein Chemistry Section, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT; and
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD
| | - Markus Berger Oliveira
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD;
| |
Collapse
|
32
|
Martin-Martin I, Valenzuela Leon PC, Amo L, Shrivastava G, Iniguez E, Aryan A, Brooks S, Kojin BB, Williams AE, Bolland S, Ackerman H, Adelman ZN, Calvo E. Aedes aegypti sialokinin facilitates mosquito blood feeding and modulates host immunity and vascular biology. Cell Rep 2022; 39:110648. [PMID: 35417706 PMCID: PMC9082008 DOI: 10.1016/j.celrep.2022.110648] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/01/2022] [Accepted: 03/17/2022] [Indexed: 11/28/2022] Open
Abstract
Saliva from mosquitoes contains vasodilators that antagonize vasoconstrictors produced at the bite site. Sialokinin is a vasodilator present in the saliva of Aedes aegypti. Here, we investigate its function and describe its mechanism of action during blood feeding. Sialokinin induces nitric oxide release similar to substance P. Sialokinin-KO mosquitoes produce lower blood perfusion than parental mosquitoes at the bite site during probing and have significantly longer probing times, which result in lower blood feeding success. In contrast, there is no difference in feeding between KO and parental mosquitoes when using artificial membrane feeders or mice that are treated with a substance P receptor antagonist, confirming that sialokinin interferes with host hemostasis via NK1R signaling. While sialokinin-KO saliva does not affect virus infection in vitro, it stimulates macrophages and inhibits leukocyte recruitment in vivo. This work highlights the biological functionality of salivary proteins in blood feeding.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Laura Amo
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Eva Iniguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Azadeh Aryan
- Department of Entomology and Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Steven Brooks
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Bianca B Kojin
- Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Adeline E Williams
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins 80523, CO, USA
| | - Silvia Bolland
- Department of Entomology and Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Zach N Adelman
- Department of Entomology and Fralin Life Science Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA; Department of Entomology, Texas A&M University, College Station, TX 77843, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
33
|
Sri-In C, Thontiravong A, Bartholomay LC, Tiawsirisup S. Effects of Aedes aegypti salivary protein on duck Tembusu virus replication and transmission in salivary glands. Acta Trop 2022; 228:106310. [PMID: 35032469 DOI: 10.1016/j.actatropica.2022.106310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/29/2022]
Abstract
Duck Tembusu virus (DTMUV) infection is an arthropod-borne viral disease that affects many poultry species, including ducks, chickens, and geese. Aedes aegypti mosquito is an important vector of DTMUV. This study sought to determine whether any individual Ae. aegypti salivary protein modulated DTMUV replication in the mosquito salivary gland. Ae. aegypti salivary gland protein of 34 kDa (AaSG34) was found to be expressed explicitly in mosquito salivary glands and was upregulated following DTMUV infection. Thus, AaSG34 was silenced in mosquitoes via RNA interference using double strand RNA (dsRNA), and the mosquitoes were then infected with DTMUV to elucidate their effects on DTMUV replication and transmission. Transcripts of the DTMUV genome in salivary glands and virus titer in saliva were significantly diminished when AaSG34 was silenced, indicating that its presence enhances DTMUV replication in the salivary glands and DTMUV dissemination to saliva. Furthermore, the expression of antimicrobial peptides (AMPs) was upregulated upon AaSG34 silenced. Our results demonstrate that AaSG34 may play a vital role in the suppression of antiviral immune responses to enhance DTMUV replication and transmission. We thus provide new information on the effect of the AaSG34 salivary protein on DTMUV replication in Ae. aegypti as the mechanism of blocking virus transmission to the host.
Collapse
Affiliation(s)
- Chalida Sri-In
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Aunyaratana Thontiravong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand; Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin, United States
| | - Sonthaya Tiawsirisup
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
34
|
Arévalo-Herrera M, Gaitán X, Larmat-Delgado M, Caicedo MA, Herrera SM, Henao-Giraldo J, Castellanos A, Devaud JC, Pannatier A, Oñate J, Corradin G, Herrera S. Randomized clinical trial to assess the protective efficacy of a Plasmodium vivax CS synthetic vaccine. Nat Commun 2022; 13:1603. [PMID: 35338131 PMCID: PMC8956637 DOI: 10.1038/s41467-022-29226-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 02/24/2022] [Indexed: 12/22/2022] Open
Abstract
A randomized, double-blind, controlled vaccine clinical trial was conducted to assess, as the primary outcome, the safety and protective efficacy of the Plasmodium vivax circumsporozoite (CS) protein in healthy malaria-naïve (phase IIa) and semi-immune (phase IIb) volunteers. Participants (n = 35) were randomly selected from a larger group (n = 121) and further divided into naïve (n = 17) and semi-immune (n = 18) groups and were immunized at months 0, 2, and 6 with PvCS formulated in Montanide ISA-51 adjuvant or placebo (adjuvant alone). Specific antibodies and IFN-γ responses to PvCS were determined as secondary outcome; all experimental volunteers developed specific IgG and IFN-γ. Three months after the last immunization, all participants were subjected to controlled human malaria infection. All naive controls became infected and drastic parasitemia reduction, including sterile protection, developed in several experimental volunteers in phase IIa (6/11) (54%, 95% CI 0.25-0.84) and phase IIb (7/11) (64%, 95% CI 0.35-0.92). However, no difference in parasitemia was observed between the phase IIb experimental and control subgroups. In conclusion, this study demonstrates significant protection in both naïve and semi-immune volunteers, encouraging further PvCS vaccine clinical development. Trial registration number NCT02083068. This trial was funded by Colciencias (grant 529-2009), NHLBI (grant RHL086488 A), and MVDC/CIV Foundation (grant 2014-1206).
Collapse
Affiliation(s)
- Myriam Arévalo-Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
- Caucaseco Scientific Research Center, Cali, Colombia
| | - Xiomara Gaitán
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
| | | | | | | | | | | | | | - André Pannatier
- Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | - Sócrates Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia.
- Caucaseco Scientific Research Center, Cali, Colombia.
| |
Collapse
|
35
|
Valenzuela-Leon PC, Shrivastava G, Martin-Martin I, Cardenas JC, Londono-Renteria B, Calvo E. Multiple Salivary Proteins from Aedes aegypti Mosquito Bind to the Zika Virus Envelope Protein. Viruses 2022; 14:v14020221. [PMID: 35215815 PMCID: PMC8876891 DOI: 10.3390/v14020221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/31/2022] Open
Abstract
Aedes aegypti mosquitoes are important vectors of several debilitating and deadly arthropod-borne (arbo) viruses, including Yellow Fever virus, Dengue virus, West Nile virus and Zika virus (ZIKV). Arbovirus transmission occurs when an infected mosquito probes the host’s skin in search of a blood meal. Salivary proteins from mosquitoes help to acquire blood and have also been shown to enhance pathogen transmission in vivo and in vitro. Here, we evaluated the interaction of mosquito salivary proteins with ZIKV by surface plasmon resonance and enzyme-linked immunosorbent assay. We found that three salivary proteins AAEL000793, AAEL007420, and AAEL006347 bind to the envelope protein of ZIKV with nanomolar affinities. Similar results were obtained using virus-like particles in binding assays. These interactions have no effect on viral replication in cultured endothelial cells and keratinocytes. Additionally, we found detectable antibody levels in ZIKV and DENV serum samples against the recombinant proteins that interact with ZIKV. These results highlight complex interactions between viruses, salivary proteins and antibodies that could be present during viral transmissions.
Collapse
Affiliation(s)
- Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (P.C.V.-L.); (G.S.); (I.M.-M.)
| | - Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (P.C.V.-L.); (G.S.); (I.M.-M.)
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (P.C.V.-L.); (G.S.); (I.M.-M.)
| | - Jenny C. Cardenas
- Arbovirology Laboratory, Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (J.C.C.); (B.L.-R.)
| | - Berlin Londono-Renteria
- Arbovirology Laboratory, Department of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (J.C.C.); (B.L.-R.)
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (P.C.V.-L.); (G.S.); (I.M.-M.)
- Correspondence:
| |
Collapse
|
36
|
Kearney EA, Agius PA, Chaumeau V, Cutts JC, Simpson JA, Fowkes FJI. Anopheles salivary antigens as serological biomarkers of vector exposure and malaria transmission: A systematic review with multilevel modelling. eLife 2021; 10:e73080. [PMID: 34939933 PMCID: PMC8860437 DOI: 10.7554/elife.73080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/21/2021] [Indexed: 12/25/2022] Open
Abstract
Background Entomological surveillance for malaria is inherently resource-intensive and produces crude population-level measures of vector exposure which are insensitive in low-transmission settings. Antibodies against Anopheles salivary proteins measured at the individual level may serve as proxy biomarkers for vector exposure and malaria transmission, but their relationship is yet to be quantified. Methods A systematic review of studies measuring antibodies against Anopheles salivary antigens (PROSPERO: CRD42020185449). Multilevel modelling (to account for multiple study-specific observations [level 1], nested within study [level 2], and study nested within country [level 3]) estimated associations between seroprevalence with Anopheles human biting rate (HBR) and malaria transmission measures. Results From 3981 studies identified in literature searches, 42 studies across 16 countries were included contributing 393 study-specific observations of anti-Anopheles salivary antibodies determined in 42,764 samples. A positive association between HBR (log transformed) and seroprevalence was found; overall a twofold (100% relative) increase in HBR was associated with a 23% increase in odds of seropositivity (OR: 1.23, 95% CI: 1.10-1.37; p<0.001). The association between HBR and Anopheles salivary antibodies was strongest with concordant, rather than discordant, Anopheles species. Seroprevalence was also significantly positively associated with established epidemiological measures of malaria transmission: entomological inoculation rate, Plasmodium spp. prevalence, and malarial endemicity class. Conclusions Anopheles salivary antibody biomarkers can serve as a proxy measure for HBR and malaria transmission, and could monitor malaria receptivity of a population to sustain malaria transmission. Validation of Anopheles species-specific biomarkers is important given the global heterogeneity in the distribution of Anopheles species. Salivary biomarkers have the potential to transform surveillance by replacing impractical, inaccurate entomological investigations, especially in areas progressing towards malaria elimination. Funding Australian National Health and Medical Research Council, Wellcome Trust.
Collapse
Affiliation(s)
- Ellen A Kearney
- The McFarlane Burnet Institute of Medical Research and Public HealthMelbourneAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of MelbourneMelbourneAustralia
| | - Paul A Agius
- The McFarlane Burnet Institute of Medical Research and Public HealthMelbourneAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of MelbourneMelbourneAustralia
- Department of Epidemiology and Preventive Medicine, Monash UniversityMelbourneAustralia
| | - Victor Chaumeau
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityMae SotThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Julia C Cutts
- The McFarlane Burnet Institute of Medical Research and Public HealthMelbourneAustralia
- Department of Medicine at the Doherty Institute, The University of MelbourneMelbourneAustralia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of MelbourneMelbourneAustralia
| | - Freya JI Fowkes
- The McFarlane Burnet Institute of Medical Research and Public HealthMelbourneAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of MelbourneMelbourneAustralia
- Department of Epidemiology and Preventive Medicine, Monash UniversityMelbourneAustralia
| |
Collapse
|
37
|
Arora G, Sajid A, Chuang YM, Dong Y, Gupta A, Gambardella K, DePonte K, Almeras L, Dimopolous G, Fikrig E. Immunomodulation by Mosquito Salivary Protein AgSAP Contributes to Early Host Infection by Plasmodium. mBio 2021; 12:e0309121. [PMID: 34903042 PMCID: PMC8669493 DOI: 10.1128/mbio.03091-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022] Open
Abstract
Malaria is caused when Plasmodium sporozoites are injected along with saliva by an anopheline mosquito into the dermis of a vertebrate host. Arthropod saliva has pleiotropic effects that can influence local host responses, pathogen transmission, and exacerbation of the disease. A mass spectrometry screen identified mosquito salivary proteins that are associated with Plasmodium sporozoites during saliva secretions. In this study, we demonstrate that one of these salivary antigens, Anopheles gambiae sporozoite-associated protein (AgSAP), interacts directly with Plasmodium falciparum and Plasmodium berghei sporozoites. AgSAP binds to heparan sulfate and inhibits local inflammatory responses in the skin. The silencing of AgSAP in mosquitoes reduces their ability to effectively transmit sporozoites to mice. Moreover, immunization with AgSAP decreases the Plasmodium burden in mice that are bitten by Plasmodium-infected mosquitoes. These data suggest that AgSAP facilitates early Plasmodium infection in the vertebrate host and serves as a target for the prevention of malaria. IMPORTANCE Malaria is a vector-borne disease caused by Plasmodium sporozoites. When an anopheline mosquito bites its host, it releases Plasmodium sporozoites as well as saliva components. Mosquito proteins have the potential to serve as antigens to prevent or influence malaria without directly targeting the pathogen. This may help set a new paradigm for vaccine development. In this study, we have elucidated the role of a novel salivary antigen, named Anopheles gambiae sporozoite-associated protein (AgSAP). The results presented here show that AgSAP interacts with Plasmodium falciparum and Plasmodium berghei sporozoites and modulates local inflammatory responses in the skin. Furthermore, our results show that AgSAP is a novel mosquito salivary antigen that influences the early stages of Plasmodium infection in the vertebrate host. Individuals living in countries where malaria is endemic generate antibodies against AgSAP, which indicates that AgSAP can serve as a biomarker for disease prevalence and epidemiological analysis.
Collapse
Affiliation(s)
- Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yuemei Dong
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Akash Gupta
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kristen Gambardella
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kathleen DePonte
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lionel Almeras
- Unité de Parasitologie et Entomologie, Département de Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France
- Aix Marseille Université, IRD, AP-HM, SSA, UMR Vecteurs-Infections Tropicales et Méditerranéennes (VITROME), IHU-Méditerranée Infection, Marseille, France
| | - George Dimopolous
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Influence of Host-Related Factors and Exposure to Mosquito Bites on the Dynamics of Antibody Response to Plasmodium falciparum Antigens. Trop Med Infect Dis 2021; 6:tropicalmed6040185. [PMID: 34698307 PMCID: PMC8544703 DOI: 10.3390/tropicalmed6040185] [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] [Received: 09/09/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 11/24/2022] Open
Abstract
Humoral immunity to Plasmodium falciparum is acquired after repeated infections, and can lead to clinical protection. This study aimed to evaluate how human-, parasite-, and environment-related determinants can modulate the dynamics of IgG responses to Plasmodium falciparum after an infection. Individuals (n = 68, average age = 8.2 years) with uncomplicated malaria were treated with ACT and followed up for 42 days. IgG responses to P. falciparum merozoite antigens (PfMSP1, PfMSP3, PfAMA1, PfGLURP-R0), to whole schizont extract (PfSchz), and to Anopheles gSG6-P1 and Aedes Nterm–34 kDa salivary peptides were measured. Regression analyses were used to identify factors that influence the dynamics of IgG response to P. falciparum antigen between D0 and D42, including demographic and biological factors and the level of exposure to mosquito bites. The dynamics of IgG response to P. falciparum differed according to the antigen. According to multivariate analysis, IgG responses to PfSchz and to PfGLURP-R0 appear to be affected by exposure to Aedes saliva and are associated with age, parasite density, and anti-Plasmodium pre-existing immune response at study inclusion. The present work shows that human exposure to Aedes saliva may contribute, in addition to other factors, to the regulation of anti-Plasmodium immune responses during a natural infection.
Collapse
|
39
|
Kumar S, Hol FJH, Pujhari S, Ellington C, Narayanan HV, Li H, Rasgon JL, Prakash M. A microfluidic platform for highly parallel bite by bite profiling of mosquito-borne pathogen transmission. Nat Commun 2021; 12:6018. [PMID: 34650045 PMCID: PMC8516912 DOI: 10.1038/s41467-021-26300-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/03/2021] [Indexed: 01/02/2023] Open
Abstract
Mosquito bites transmit a number of pathogens via salivary droplets deposited during blood-feeding, resulting in potentially fatal diseases. Little is known about the genomic content of these nanodroplets, including the transmission dynamics of live pathogens. Here we introduce Vectorchip, a low-cost, scalable microfluidic platform enabling high-throughput molecular interrogation of individual mosquito bites. We introduce an ultra-thin PDMS membrane which acts as a biting interface to arrays of micro-wells. Freely-behaving mosquitoes deposit saliva droplets by biting into these micro-wells. By modulating membrane thickness, we observe species-dependent differences in mosquito biting capacity, utilizable for selective sample collection. We demonstrate RT-PCR and focus-forming assays on-chip to detect mosquito DNA, Zika virus RNA, as well as quantify infectious Mayaro virus particles transmitted from single mosquito bites. The Vectorchip presents a promising approach for single-bite-resolution laboratory and field characterization of vector-pathogen communities, and could serve as a powerful early warning sentinel for mosquito-borne diseases.
Collapse
Affiliation(s)
- Shailabh Kumar
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Felix J H Hol
- Department of Bioengineering, Stanford University, Stanford, CA, USA.,Insect Virus Interactions Unit, Department of Virology, Institut Pasteur, UMR2000, CNRS, Paris, France.,Center for Research and Interdisciplinarity, U1284 INSERM, Université de Paris, Paris, France
| | - Sujit Pujhari
- Department of Entomology, The Center for Infectious Disease Dynamics, and the Huck Institutes of The Life Sciences, The Pennsylvania State University, University Park, PA, USA.,Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Clayton Ellington
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Hongquan Li
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Jason L Rasgon
- Department of Entomology, The Center for Infectious Disease Dynamics, and the Huck Institutes of The Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Manu Prakash
- Department of Bioengineering, Stanford University, Stanford, CA, USA. .,Woods Institute for the Environment, Stanford University, Stanford, CA, USA.
| |
Collapse
|
40
|
Park SL, Huang YJS, Lyons AC, Ayers VB, Hettenbach SM, McVey DS, Noronha LE, Burton KR, Hsu WW, Higgs S, Vanlandingham DL. Mosquito Saliva Modulates Japanese Encephalitis Virus Infection in Domestic Pigs. FRONTIERS IN VIROLOGY 2021. [DOI: 10.3389/fviro.2021.724016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that is the leading cause of pediatric viral encephalitis in Asia. Japanese encephalitis virus is transmitted by Culex species mosquitoes that also vector several zoonotic flaviviruses. Despite the knowledge that mosquito saliva contains molecules that may alter flavivirus pathogenesis, whether or not the deposition of viruses by infected mosquitoes has an impact on the kinetics and severity of JEV infection has not been thoroughly examined, especially in mammalian species involved in the enzootic transmission. Most JEV pathogenesis models were established using needle inoculation. Mouse models for West Nile (WNV) and dengue (DENV) viruses have shown that mosquito saliva can potentiate flavivirus infections and exacerbate disease symptoms. In this study, we determined the impact of mosquito salivary components on the pathogenesis of JEV in pigs, a species directly involved in its transmission cycle as an amplifying host. Interestingly, co-injection of JEV and salivary gland extract (SGE) collected from Culex quinquefasciatus produced milder febrile illness and shortened duration of nasal shedding but had no demonstrable impact on viremia and neuroinvasion. Our findings highlight that mosquito salivary components can differentially modulate the outcomes of flavivirus infections in amplifying hosts and in mouse models.
Collapse
|
41
|
Duffy FJ, Du Y, Carnes J, Epstein JE, Hoffman SL, Abdulla S, Jongo S, Mpina M, Daubenberger C, Aitchison JD, Stuart K. Early whole blood transcriptional responses to radiation-attenuated Plasmodium falciparum sporozoite vaccination in malaria naïve and malaria pre-exposed adult volunteers. Malar J 2021; 20:308. [PMID: 34243763 PMCID: PMC8267772 DOI: 10.1186/s12936-021-03839-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/29/2021] [Indexed: 12/03/2022] Open
Abstract
Background Vaccination with radiation-attenuated Plasmodium falciparum sporozoites is known to induce protective immunity. However, the mechanisms underlying this protection remain unclear. In this work, two recent radiation-attenuated sporozoite vaccination studies were used to identify potential transcriptional correlates of vaccination-induced protection. Methods Longitudinal whole blood RNAseq transcriptome responses to immunization with radiation-attenuated P. falciparum sporozoites were analysed and compared across malaria-naïve adult participants (IMRAS) and malaria-experienced adult participants (BSPZV1). Parasite dose and method of delivery differed between trials, and immunization regimens were designed to achieve incomplete protective efficacy. Observed protective efficacy was 55% in IMRAS and 20% in BSPZV1. Study vaccine dosings were chosen to elicit both protected and non-protected subjects, so that protection-associated responses could be identified. Results Analysis of comparable time points up to 1 week after the first vaccination revealed a shared cross-study transcriptional response programme, despite large differences in number and magnitude of differentially expressed genes between trials. A time-dependent regulatory programme of coherent blood transcriptional modular responses was observed, involving induction of inflammatory responses 1–3 days post-vaccination, with cell cycle responses apparent by day 7 in protected individuals from both trials. Additionally, strongly increased induction of inflammation and interferon-associated responses was seen in non-protected IMRAS participants. All individuals, except for non-protected BSPZV1 participants, showed robust upregulation of cell-cycle associated transcriptional responses post vaccination. Conclusions In summary, despite stark differences between the two studies, including route of vaccination and status of malaria exposure, responses were identified that were associated with protection after PfRAS vaccination. These comprised a moderate early interferon response peaking 2 days post vaccination, followed by a later proliferative cell cycle response steadily increasing over the first 7 days post vaccination. Non-protection is associated with deviations from this model, observed in this study with over-induction of early interferon responses in IMRAS and failure to mount a cell cycle response in BSPZV1. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03839-3.
Collapse
Affiliation(s)
- Fergal J Duffy
- Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, WA, USA.
| | - Ying Du
- Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, WA, USA
| | - Jason Carnes
- Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, WA, USA
| | - Judith E Epstein
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, USA
| | | | | | - Said Jongo
- Ifakara Health Institute, Bagamoyo, Tanzania
| | - Maxmillian Mpina
- Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.,University of Basel, Petersplatz 1, 4001, Basel, Switzerland.,Ifakara Health Institute, Bagamoyo, Tanzania
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, 4002, Basel, Switzerland.,University of Basel, Petersplatz 1, 4001, Basel, Switzerland
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, WA, USA
| | - Ken Stuart
- Center for Global Infectious Disease Research, Seattle Children's Hospital, Seattle, WA, USA.
| |
Collapse
|
42
|
Blight J, Sala KA, Atcheson E, Kramer H, El-Turabi A, Real E, Dahalan FA, Bettencourt P, Dickinson-Craig E, Alves E, Salman AM, Janse CJ, Ashcroft FM, Hill AV, Reyes-Sandoval A, Blagborough AM, Baum J. Dissection-independent production of Plasmodium sporozoites from whole mosquitoes. Life Sci Alliance 2021; 4:e202101094. [PMID: 34135099 PMCID: PMC8321652 DOI: 10.26508/lsa.202101094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/05/2023] Open
Abstract
Progress towards a protective vaccine against malaria remains slow. To date, only limited protection has been routinely achieved following immunisation with either whole-parasite (sporozoite) or subunit-based vaccines. One major roadblock to vaccine progress, and to pre-erythrocytic parasite biology in general, is the continued reliance on manual salivary gland dissection for sporozoite isolation from infected mosquitoes. Here, we report development of a multi-step method, based on batch processing of homogenised whole mosquitoes, slurry, and density-gradient filtration, which combined with free-flow electrophoresis rapidly produces a pure, infective sporozoite inoculum. Human-infective Plasmodium falciparum and rodent-infective Plasmodium berghei sporozoites produced in this way are two- to threefold more infective than salivary gland dissection sporozoites in in vitro hepatocyte infection assays. In an in vivo rodent malaria model, the same P. berghei sporozoites confer sterile protection from mosquito-bite challenge when immunisation is delivered intravenously or 60-70% protection when delivered intramuscularly. By improving purity, infectivity, and immunogenicity, this method represents a key advancement in capacity to produce research-grade sporozoites, which should impact delivery of a whole-parasite based malaria vaccine at scale in the future.
Collapse
Affiliation(s)
- Joshua Blight
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Katarzyna A Sala
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
| | - Erwan Atcheson
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Holger Kramer
- Department of Physiology, Anatomy and Genetics, Henry Wellcome Building for Gene Function, University of Oxford, Oxford, UK
- Medical Research Council London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Aadil El-Turabi
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Eliana Real
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
| | - Farah A Dahalan
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
| | - Paulo Bettencourt
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Emma Dickinson-Craig
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Eduardo Alves
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Ahmed M Salman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Chris J Janse
- Department of Parasitology, Leiden Malaria Research Group, Center of Infectious Diseases, Leiden University Medical Center, (LUMC, L4-Q), Leiden, The Netherlands
| | - Frances M Ashcroft
- Department of Physiology, Anatomy and Genetics, Henry Wellcome Building for Gene Function, University of Oxford, Oxford, UK
| | - Adrian Vs Hill
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, UK
- Instituto Politécnico Nacional, Mexico City, Mexico
| | - Andrew M Blagborough
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, London, UK
| |
Collapse
|
43
|
Schäfer C, Zanghi G, Vaughan AM, Kappe SHI. Plasmodium vivax Latent Liver Stage Infection and Relapse: Biological Insights and New Experimental Tools. Annu Rev Microbiol 2021; 75:87-106. [PMID: 34196569 DOI: 10.1146/annurev-micro-032421-061155] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium vivax is the most widespread human malaria parasite, in part because it can form latent liver stages known as hypnozoites after transmission by female anopheline mosquitoes to human hosts. These persistent stages can activate weeks, months, or even years after the primary clinical infection; replicate; and initiate relapses of blood stage infection, which causes disease and recurring transmission. Eliminating hypnozoites is a substantial obstacle for malaria treatment and eradication since the hypnozoite reservoir is undetectable and unaffected by most antimalarial drugs. Importantly, in some parts of the globe where P. vivax malaria is endemic, as many as 90% of P. vivax blood stage infections are thought to be relapses rather than primary infections, rendering the hypnozoite a major driver of P. vivax epidemiology. Here, we review the biology of the hypnozoite and recent discoveries concerning this enigmatic parasite stage. We discuss treatment and prevention challenges, novel animal models to study hypnozoites and relapse, and hypotheses related to hypnozoite formation and activation. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.,Deparment of Global Health, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
44
|
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: 0.8] [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
|
45
|
Complex Roles of Neutrophils during Arboviral Infections. Cells 2021; 10:cells10061324. [PMID: 34073501 PMCID: PMC8227388 DOI: 10.3390/cells10061324] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Arboviruses are known to cause large-scale epidemics in many parts of the world. These arthropod-borne viruses are a large group consisting of viruses from a wide range of families. The ability of their vector to enhance viral pathogenesis and transmission makes the development of treatments against these viruses challenging. Neutrophils are generally the first leukocytes to be recruited to a site of infection, playing a major role in regulating inflammation and, as a result, viral replication and dissemination. However, the underlying mechanisms through which neutrophils control the progression of inflammation and disease remain to be fully understood. In this review, we highlight the major findings from recent years regarding the role of neutrophils during arboviral infections. We discuss the complex nature of neutrophils in mediating not only protection, but also augmenting disease pathology. Better understanding of neutrophil pathways involved in effective protection against arboviral infections can help identify potential targets for therapeutics.
Collapse
|
46
|
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
|
47
|
Trammell CE, Goodman AG. Host Factors That Control Mosquito-Borne Viral Infections in Humans and Their Vector. Viruses 2021; 13:748. [PMID: 33923307 PMCID: PMC8145797 DOI: 10.3390/v13050748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Mosquito-borne viral infections are responsible for a significant degree of morbidity and mortality across the globe due to the severe diseases these infections cause, and they continue to increase each year. These viruses are dependent on the mosquito vector as the primary means of transmission to new vertebrate hosts including avian, livestock, and human populations. Due to the dynamic host environments that mosquito-borne viruses pass through as they are transmitted between vector and vertebrate hosts, there are various host factors that control the response to infection over the course of the pathogen's life cycle. In this review, we discuss these host factors that are present in either vector or vertebrate models during infection, how they vary or are conserved between hosts, and their implications in future research pertaining to disease prevention and treatment.
Collapse
Affiliation(s)
- Chasity E. Trammell
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA;
- NIH Protein Biotechnology Training Program, Washington State University, Pullman, WA 99164-6240, USA
| | - Alan G. Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99163, USA;
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
48
|
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: 0.5] [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
|
49
|
Role of cytokines produced by T helper immune-modulators in dengue pathogenesis: A systematic review and meta-analysis. Acta Trop 2021; 216:105823. [PMID: 33421421 DOI: 10.1016/j.actatropica.2021.105823] [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] [Received: 05/07/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVES Modulation of the immune reaction is essential in the development of various diseases, including dengue's "Cytokine Tsunami", an increase in vascular permeability with concomitant severe vascular leakage. We aim to identify the role of T-helper (Th) cells, Th2 and Th7, with their related cytokines in dengue pathogenesis. MATERIAL AND METHODS Nine electronic databases and manual search were applied to detect available publications. A meta-analysis using a fixed- or random-effect model was performed to measure standardized mean difference (SMD) with 95% confidence interval (CI). The National Institute of Health (NIH) tools for observational cohort, cross-sectional, and case-control studies were used to examine the risk of bias. The protocol was recorded in PROSPERO with CRD42017060230. RESULTS A total of 38 articles were found including 19 case-control, 11 cross-sectional and 8 prospective cohort studies. We indicated that Th2 cytokines (IL-4, IL-6, IL-8) and Th17 cytokine (IL-17) in dengue patients were notably higher than in a healthy control group in acute phase (SMD = 1.59, 95% CI [0.68, 2.51], p = 0.001; SMD = 1.24, 95% CI [0.41, 2.06], p = 0.003; SMD = 1.13, 95% CI [0.61, 1.66], p<0.0001; SMD = 1.74, 95% CI [0.87, 2.61], p<0.0001), respectively. CONCLUSIONS This study provides evidence of the significant roles of IL-4, IL-6, IL-8, IL-10 and IL-17 in the pathogenesis of developing a severe reaction in dengue fever. However, to fully determine the association of Th cytokines with dengue, it is necessary to perform further studies to assess kinetic levels during the duration of the illness.
Collapse
|
50
|
Kern O, Valenzuela Leon PC, Gittis AG, Bonilla B, Cruz P, Chagas AC, Ganesan S, Ribeiro JMC, Garboczi DN, Martin-Martin I, Calvo E. The structures of two salivary proteins from the West Nile vector Culex quinquefasciatus reveal a beta-trefoil fold with putative sugar binding properties. Curr Res Struct Biol 2021; 3:95-105. [PMID: 34235489 PMCID: PMC8244437 DOI: 10.1016/j.crstbi.2021.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/22/2023] Open
Abstract
Female mosquitoes require blood meals for egg development. The saliva of blood feeding arthropods contains biochemically active molecules, whose anti-hemostatic and anti-inflammatory properties facilitate blood feeding on vertebrate hosts. While transcriptomics has presented new opportunities to investigate the diversity of salivary proteins from hematophagous arthropods, many of these proteins remain functionally undescribed. Previous transcriptomic analysis of female salivary glands from Culex quinquefasciatus, an important vector of parasitic and viral infections, uncovered a 12-member family of putatively secreted proteins of unknown function, named the Cysteine and Tryptophan-Rich (CWRC) proteins. Here, we present advances in the characterization of two C. quinquefasciatus CWRC family members, CqDVP-2 and CqDVP-4, including their enrichment in female salivary glands, their specific localization within salivary gland tissues, evidence that these proteins are secreted into the saliva, and their native crystal structures, at 2.3 Å and 1.87 Å, respectively. The β-trefoil fold common to CqDVP-2 and CqDVP-4 is similar to carbohydrate-binding proteins, including the B subunit of the AB toxin, ricin, from the castor bean Ricinus communis. Further, we used a glycan array approach, which identifies carbohydrate ligands associated with inflammatory processes and signal transduction. Glycan array 300 testing identified 100 carbohydrate moieties with positive binding to CqDVP-2, and 77 glycans with positive binding to CqDVP-4. The glycan with the highest relative fluorescence intensities, which exhibited binding to both CqDVP-2 and CqDVP-4, was used for molecular docking experiments. We hypothesize that these proteins bind to carbohydrates on the surface of cells important to host immunology. Given that saliva is deposited into the skin during a mosquito bite, and acts as the vehicle for arbovirus inoculation, understanding the role of these proteins in pathogen transmission is of critical importance. This work presents the first solved crystal structures of C. quinquefasciatus salivary proteins with unknown function. These two molecules are the second and third structures reported from salivary proteins from C. quinquefasciatus, an important, yet understudied disease vector.
Collapse
Affiliation(s)
- Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Apostolos G Gittis
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Brian Bonilla
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Phillip Cruz
- Bioinformatics and Computational Biosciences Branch. Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Jose M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - David N Garboczi
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| |
Collapse
|