1
|
Chi Y, Zhang H, Chen S, Cheng Y, Zhang X, Jia D, Chen Q, Chen H, Wei T. Leafhopper salivary carboxylesterase suppresses JA-Ile synthesis to facilitate initial arbovirus transmission in rice phloem. PLANT COMMUNICATIONS 2024; 5:100939. [PMID: 38725245 PMCID: PMC11412928 DOI: 10.1016/j.xplc.2024.100939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/16/2024] [Accepted: 05/01/2024] [Indexed: 06/09/2024]
Abstract
Plant jasmonoyl-L-isoleucine (JA-Ile) is a major defense signal against insect feeding, but whether or how insect salivary effectors suppress JA-Ile synthesis and thus facilitate viral transmission in the plant phloem remains elusive. Insect carboxylesterases (CarEs) are the third major family of detoxification enzymes. Here, we identify a new leafhopper CarE, CarE10, that is specifically expressed in salivary glands and is secreted into the rice phloem as a saliva component. Leafhopper CarE10 directly binds to rice jasmonate resistant 1 (JAR1) and promotes its degradation by the proteasome system. Moreover, the direct association of CarE10 with JAR1 clearly impairs JAR1 enzyme activity for conversion of JA to JA-Ile in an in vitro JA-Ile synthesis system. A devastating rice reovirus activates and promotes the co-secretion of virions and CarE10 via virus-induced vesicles into the saliva-storing salivary cavities of the leafhopper vector and ultimately into the rice phloem to establish initial infection. Furthermore, a virus-mediated increase in CarE10 secretion or overexpression of CarE10 in transgenic rice plants causes reduced levels of JAR1 and thus suppresses JA-Ile synthesis, promoting host attractiveness to insect vectors and facilitating initial viral transmission. Our findings provide insight into how the insect salivary protein CarE10 suppresses host JA-Ile synthesis to promote initial virus transmission in the rice phloem.
Collapse
Affiliation(s)
- Yunhua Chi
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hongxiang Zhang
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Siyu Chen
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Yu Cheng
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Xiaofeng Zhang
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Dongsheng Jia
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Qian Chen
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hongyan Chen
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Taiyun Wei
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| |
Collapse
|
2
|
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 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
|
3
|
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
|
4
|
Wang Z, Nie K, Liang Y, Niu J, Yu X, Zhang O, Liu L, Shi X, Wang Y, Feng X, Zhu Y, Wang P, Cheng G. A mosquito salivary protein-driven influx of myeloid cells facilitates flavivirus transmission. EMBO J 2024; 43:1690-1721. [PMID: 38378891 PMCID: PMC11066113 DOI: 10.1038/s44318-024-00056-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/22/2024] Open
Abstract
Mosquitoes transmit many disease-relevant flaviviruses. Efficient viral transmission to mammalian hosts requires mosquito salivary factors. However, the specific salivary components facilitating viral transmission and their mechanisms of action remain largely unknown. Here, we show that a female mosquito salivary gland-specific protein, here named A. aegypti Neutrophil Recruitment Protein (AaNRP), facilitates the transmission of Zika and dengue viruses. AaNRP promotes a rapid influx of neutrophils, followed by virus-susceptible myeloid cells toward mosquito bite sites, which facilitates establishment of local infection and systemic dissemination. Mechanistically, AaNRP engages TLR1 and TLR4 of skin-resident macrophages and activates MyD88-dependent NF-κB signaling to induce the expression of neutrophil chemoattractants. Inhibition of MyD88-NF-κB signaling with the dietary phytochemical resveratrol reduces AaNRP-mediated enhancement of flavivirus transmission by mosquitoes. These findings exemplify how salivary components can aid viral transmission, and suggest a potential prophylactic target.
Collapse
Affiliation(s)
- Zhaoyang Wang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Kaixiao Nie
- Department of Pathogen Biology, School of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yan Liang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jichen Niu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Xi Yu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Oujia Zhang
- Department of Pathogen Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100086, China
| | - Long Liu
- Institute of Virology, Hubei University of Medicine, Shiyan, 442000, China
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Xiaolu Shi
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Yibaina Wang
- China National Center for Food Safety Risk Assessment, Beijing, 100022, China
| | - Xuechun Feng
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
| | - Yibin Zhu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China.
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
- Southwest United Graduate School, Kunming, 650092, China.
| |
Collapse
|
5
|
Keskek Turk Y, Barningham LD, McKimmie CS. Sensing the danger in mosquito spit. EMBO J 2024; 43:1687-1689. [PMID: 38499785 PMCID: PMC11065867 DOI: 10.1038/s44318-024-00073-w] [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: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Recent study identifies AaNRP as an arboviral infection-promoting factor in Aedes aegypti mosquito saliva that promotes recruitment of virus-susceptible myeloid cells.
Collapse
Affiliation(s)
- Yonca Keskek Turk
- Virus Host Interaction Team, School of Medicine, University of Leeds, Leeds, UK
| | - Liam D Barningham
- Virus Host Interaction Team, School of Medicine, University of Leeds, Leeds, UK
| | - Clive S McKimmie
- Virus Host Interaction Team, School of Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
6
|
Pei L, Hickman HD. T Cell Surveillance during Cutaneous Viral Infections. Viruses 2024; 16:679. [PMID: 38793562 PMCID: PMC11126121 DOI: 10.3390/v16050679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/26/2024] Open
Abstract
The skin is a complex tissue that provides a strong physical barrier against invading pathogens. Despite this, many viruses can access the skin and successfully replicate in either the epidermal keratinocytes or dermal immune cells. In this review, we provide an overview of the antiviral T cell biology responding to cutaneous viral infections and how these responses differ depending on the cellular targets of infection. Much of our mechanistic understanding of T cell surveillance of cutaneous infection has been gained from murine models of poxvirus and herpesvirus infection. However, we also discuss other viral infections, including flaviviruses and papillomaviruses, in which the cutaneous T cell response has been less extensively studied. In addition to the mechanisms of successful T cell control of cutaneous viral infection, we highlight knowledge gaps and future directions with possible impact on human health.
Collapse
Affiliation(s)
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
7
|
Loh SN, Anthony IR, Gavor E, Lim XS, Kini RM, Mok YK, Sivaraman J. Recognition of Aedes aegypti Mosquito Saliva Protein LTRIN by the Human Receptor LTβR for Controlling the Immune Response. BIOLOGY 2024; 13:42. [PMID: 38248473 PMCID: PMC10813304 DOI: 10.3390/biology13010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
Salivary proteins from mosquitoes have received significant attention lately due to their potential to develop therapeutic treatments or vaccines for mosquito-borne diseases. Here, we report the characterization of LTRIN (lymphotoxin beta receptor inhibitor), a salivary protein known to enhance the pathogenicity of ZIKV by interrupting the LTβR-initiated NF-κB signaling pathway and, therefore, diminish the immune responses. We demonstrated that the truncated C-terminal LTRIN (ΔLTRIN) is a dimeric protein with a stable alpha helix-dominant secondary structure, which possibly aids in withstanding the temperature fluctuations during blood-feeding events. ΔLTRIN possesses two Ca2+ binding EF-hand domains, with the second EF-hand motif playing a more significant role in interacting with LTβR. Additionally, we mapped the primary binding regions of ΔLTRIN on LTβR using hydrogen-deuterium exchange mass spectrometry (HDX-MS) and identified that 91QEKAHIAEHMDVPIDTSKMSEQELQFHY118 from the N-terminal of ΔLTRIN is the major interacting region. Together, our studies provide insight into the recognition of LTRIN by LTβR. This finding may aid in a future therapeutic and transmission-blocking vaccine development against ZIKV.
Collapse
Affiliation(s)
- Su Ning Loh
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| | - Ian Russell Anthony
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| | - Edem Gavor
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| | - Xin Shan Lim
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| | - R. Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Yu Keung Mok
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| | - J. Sivaraman
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore; (S.N.L.)
| |
Collapse
|
8
|
Fan SH, Chang Y, Xiong XY, Xiang M, Yuan WL, Yang XQ, Wei WH, Chen L, Cheng MN, Zhu FH, He SJ, Zuo JP, Lin ZM. Reversible SAHH inhibitor ameliorates MIA-induced osteoarthritis of rats through suppressing MEK/ERK pathway. Biomed Pharmacother 2024; 170:115975. [PMID: 38070246 DOI: 10.1016/j.biopha.2023.115975] [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: 09/10/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Osteoarthritis (OA) is characterized by gradual articular cartilage degradation, accompanied by persistent low-grade joint inflammation, correlating with radiographic and pain-related progression. The latent therapeutic potential of DZ2002, a reversible inhibitor of S-adenosyl-L-homocysteine hydrolase (SAHH), holds promise for OA intervention. This study endeavored to examine the therapeutic efficacy of DZ2002 within the milieu of OA. The cytotoxicity of DZ2002 was evaluated using the MTT assay on bone marrow-derived macrophages. The inhibitory impact of DZ2002 during the process of osteoclastogenesis was assessed using TRAP staining, analysis of bone resorption pits, and F-actin ring formation. Mechanistic insights were derived from qPCR and Western blot analyses. Through the intra-articular injection of monosodium iodoacetate (MIA), an experimental rat model of OA was successfully instituted. This was subsequently accompanied by a series of assessments including Von Frey filament testing, analysis of weight-bearing behaviors, and micro-CT imaging, all aimed at assessing the effectiveness of DZ2002. The findings emphasized the effectiveness of DZ2002 in mitigating osteoclastogenesis induced by M-CSF/RANKL, evident through a reduction in TRAP-positive OCs and bone resorption. Moreover, DZ2002 modulated bone resorption-associated gene and protein expression (CTSK, CTR, Integrin β3) via the MEK/ERK pathway. Encouragingly, DZ2002 also alleviates MIA-induced pain, cartilage degradation, and bone loss. In conclusion, DZ2002 emerges as a potential therapeutic contender for OA, as evidenced by its capacity to hinder in vitro M-CSF/RANKL-induced osteoclastogenesis and mitigate in vivo osteoarthritis progression. This newfound perspective provides substantial support for considering DZ2002 as a compelling agent for osteoarthritis intervention.
Collapse
Affiliation(s)
- Shu-Hui Fan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuan Chang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao-Yu Xiong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mai Xiang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Long Yuan
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen-Hui Wei
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Chen
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meng-Nan Cheng
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Ping Zuo
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China; Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Zhu Y, Liu J, Cheng G. Progress towards research on mosquito-borne arboviral transmission and infection. Sci Bull (Beijing) 2023; 68:2884-2888. [PMID: 37940452 DOI: 10.1016/j.scib.2023.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Affiliation(s)
- Yibin Zhu
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jianying Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Gong Cheng
- Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518000, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.
| |
Collapse
|
10
|
Huang Q, Gavor E, Tulsian NK, Fan J, Lin Q, Mok YK, Kini RM, Sivaraman J. Structural and functional characterization of Aedes aegypti pupal cuticle protein that controls dengue virus infection. Protein Sci 2023; 32:e4761. [PMID: 37593853 PMCID: PMC10510476 DOI: 10.1002/pro.4761] [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: 06/08/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
The pupal cuticle protein from Aedes aegypti (AaPC) inhibits dengue virus (DENV) infection; however, the underlying mechanism of this inhibition remains unknown. Here, we report that AaPC is an intrinsically disordered protein and interacts with domain I/II of the DENV envelope protein via residues Asp59, Asp61, Glu71, Asp73, Ser75, and Asp80. AaPC can directly bind to and cause the aggregation of DENV, which in turn blocks virus infection during the virus-cell fusion stage. AaPC may also influence viral recognition and attachment by interacting with human immune receptors DC-SIGN and CD4. These findings enhance our understanding of the role of AaPC in mitigating viral infection and suggest that AaPC is a potential target for developing inhibitors or antibodies to control dengue virus infection.
Collapse
Affiliation(s)
- Qingqing Huang
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Edem Gavor
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Nikhil Kumar Tulsian
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
- Department of BiochemistryNational University of SingaporeSingaporeSingapore
| | - Jingsong Fan
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Qingsong Lin
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Yu Keung Mok
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - R. Manjunatha Kini
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - J. Sivaraman
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| |
Collapse
|
11
|
Lin Z, Cheng J, Mu X, Kuang X, Li Z, Wu J. A C-type lectin in saliva of Aedes albopictus (Diptera: Culicidae) bind and agglutinate microorganisms with broad spectrum. JOURNAL OF INSECT SCIENCE (ONLINE) 2023; 23:1. [PMID: 37399114 DOI: 10.1093/jisesa/iead043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/04/2023] [Accepted: 05/28/2023] [Indexed: 07/05/2023]
Abstract
Via complex salivary mixture, mosquitos can intervene immune response and be helpful to transmit several viruses causing deadly human diseases. Some C-type lectins (CTLs) of mosquito have been reported to be pattern recognition receptor to either resist or promote pathogen invading. Here, we investigated the expression profile and agglutination function of an Aedes albopictus CTL (Aalb_CTL2) carrying a single carbohydrate-recognition domain (CRD) and WND/KPD motifs. The results showed that Aalb_CTL2 was found to be specifically expressed in mosquito saliva gland and its expression was not induced by blood-feeding. The recombinant Aalb_CTL2 (rAalb_CTL2) could agglutinate mouse erythrocytes in the presence of calcium and the agglutinating activity could be inhibited by EDTA. rAalb_CTL2 also displayed the sugar binding ability to D-mannose, D-galactose, D-glucose, and maltose. Furthermore, it was demonstrated that rAalb_CTL2 could bind and agglutinate Gram positive bacteria Staphylococcus aureus and Bacillus subtilis, Gram negative bacteria Escherichia coli and Pseudomonas aeruginosa, as well as fungus Candida albicans in vitro in a calcium dependent manner. However, rAalb_CTL2 could not promote type 2 dengue virus (DENV-2) replication in THP-1 and BHK-21 cell lines. These findings uncover that Aalb_CTL2 might be involved in the innate immunity of mosquito to resist microorganism multiplication in sugar and blood meals to help mosquito survive in the varied natural environment.
Collapse
Affiliation(s)
- Zimin Lin
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Parasitology, Guizhou Medical University, Guiyang 550025, China
| | - Jinzhi Cheng
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Parasitology, Guizhou Medical University, Guiyang 550025, China
| | - Xiaohui Mu
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Parasitology, Guizhou Medical University, Guiyang 550025, China
| | - Xiaoyuan Kuang
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Parasitology, Guizhou Medical University, Guiyang 550025, China
| | - Zhiqiang Li
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Immunology, Guizhou Medical University, Guiyang 550025, China
| | - Jiahong Wu
- The Key and Characteristic Laboratory of Modern Pathogen Biology, College of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
- Department of Parasitology, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
12
|
Cerqueira ARA, Rodrigues L, Coavoy-Sánchez SA, Teixeira SA, Feitosa KB, Taniguchi EY, Lopes LR, Cassola AC, Muscará MN, Sá-Nunes A, Costa SKP. Aedes aegypti salivary gland extract alleviates acute itching by blocking TRPA1 channels. Front Physiol 2023; 14:1055706. [PMID: 37441000 PMCID: PMC10333701 DOI: 10.3389/fphys.2023.1055706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Aedes aegypti (Ae. aegypti) saliva induces a variety of anti-inflammatory and immunomodulatory activities. Interestingly, although it is known that mosquito bites cause allergic reactions in sensitised hosts, the primary exposure of humans to Ae. aegypti does not evoke significant itching. Whether active components in the saliva of Ae. aegypti can counteract the normal itch reaction to injury produced by a histaminergic or non-histaminergic pathway in vertebrate hosts is unknown. This study investigated the effects of Ae. aegypti mosquito salivary gland extract (SGE) on sensitive reactions such as itching and associated skin inflammation. Acute pruritus and plasma extravasation were induced in mice by the intradermal injection of either compound 48/80 (C48/80), the Mas-related G protein-coupled receptor (Mrgpr) agonist chloroquine (CQ), or the transient receptor potential ankyrin 1 (TRPA1) agonist allyl isothiocyanate (AITC). The i.d. co-injection of Ae. aegypti SGE inhibited itching, plasma extravasation, and neutrophil influx evoked by C48/80, but it did not significantly affect mast cell degranulation in situ or in vitro. Additionally, SGE partially reduced CQ- and AITC-induced pruritus in vivo, suggesting that SGE affects pruriceptive nerve firing independently of the histaminergic pathway. Activation of TRPA1 significantly increased intracellular Ca2+ in TRPA-1-transfected HEK293t lineage, which was attenuated by SGE addition. We showed for the first time that Ae. aegypti SGE exerts anti-pruriceptive effects, which are partially regulated by the histamine-independent itch TRPA1 pathway. Thus, SGE may possess bioactive molecules with therapeutic potential for treating nonhistaminergic itch.
Collapse
Affiliation(s)
- Anderson R. A. Cerqueira
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Leandro Rodrigues
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | | | - Simone A. Teixeira
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Karla B. Feitosa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Erika Y. Taniguchi
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Lucia R. Lopes
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Antônio C. Cassola
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo N. Muscará
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Brazil
| | - Soraia K. P. Costa
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Wang L, Sun F, Hu J, Zuo W, Zheng Y, Wu Y, Kwok HF, Cao Z. The tick saliva peptide HIDfsin2 promotes the tick-borne virus SFTSV replication in vitro by enhancing p38 signal pathway. Arch Toxicol 2023; 97:1783-1794. [PMID: 37148319 PMCID: PMC10163292 DOI: 10.1007/s00204-023-03515-2] [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: 03/04/2023] [Accepted: 04/26/2023] [Indexed: 05/08/2023]
Abstract
Pathogens co-evolved with ticks to facilitate blood collection and pathogen transmission. Although tick saliva was recently found to be rich in bioactive peptides, it is still elusive which saliva peptide promotes virus transmission and which pathways are invovled. Here, we used a saliva peptide HIDfsin2 and a severe fever with thrombocytopenia syndrome virus (SFTSV) both carried by the tick Haemaphysalis longicornis to elucidate the relationship between tick saliva components and tick-borne viruses. HIDfsin2 was found to promote the replication of SFTSV in a dose-dependent manner in vitro. HIDfsin2 was further revealed to MKK3/6-dependently magnify the activation of p38 MAPK. The overexpression, knockdown and phosphorylation site mutation of p38α indicated that p38 MAPK activation facilitated SFTSV infection in A549 cells. Moreover, the blockade of p38 MAPK activation significantly suppressed SFTSV replication. Differently, HIDfsin2 or pharmacological inhibition of p38 MAPK activation had no effect on a mosquito-borne Zika virus (ZIKV). All these results showed that HIDfsin2 specifically promoted SFTSV replication through the MKK3/6-dependent enhancement of p38 MAPK activation. Our study provides a new perspective on the transmission of tick-borne viruses under natural conditions, and supports that the blockade of p38 MAPK activation can be a promising strategy against the mortal tick-borne virus SFTSV.
Collapse
Affiliation(s)
- Luyao Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Fang Sun
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hubei University of Medicine, Shiyan, 442000, China
| | - Jing Hu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Weimin Zuo
- Department of Biomedical Sciences, Faculty of Health Sciences, MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Yi Zheng
- Center for Medical Experiments (CME), University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518106, China
| | - Yingliang Wu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China.
| | - Zhijian Cao
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
14
|
Mancini MV, Tandavanitj R, Ant TH, Murdochy SM, Gingell DD, Setthapramote C, Natsrita P, Kohl A, Sinkins SP, Patel AH, De Lorenzo G. Evaluation of an Engineered Zika Virus-Like Particle Vaccine Candidate in a Mosquito-Mouse Transmission Model. mSphere 2023; 8:e0056422. [PMID: 36840596 PMCID: PMC10117074 DOI: 10.1128/msphere.00564-22] [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: 12/02/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
The primary route of Zika virus (ZIKV) transmission is through the bite of an infected Aedes mosquito, when it probes the skin of a vertebrate host during a blood meal. Viral particles are injected into the bite site together with mosquito saliva and a complex mixture of other components. Some of them are known to play a key role in the augmentation of the arbovirus infection in the host, with increased viremia and/or morbidity. This vector-derived contribution to the infection is not usually considered when vaccine candidates are tested in preclinical animal models. In this study, we performed a preclinical validation of a promising ZIKV vaccine candidate in a mosquito-mouse transmission model using both Asian and African ZIKV lineages. Mice were immunized with engineered ZIKV virus-like particles and subsequently infected through the bite of ZIKV-infected Aedes aegypti mosquitoes. Despite a mild increase in viremia in mosquito-infected mice compared to those infected through traditional needle injection, the vaccine protected the animals from developing the disease and strongly reduced viremia. In addition, during peak viremia, naive mosquitoes were allowed to feed on infected vaccinated and nonvaccinated mice. Our analysis of viral titers in mosquitos showed that the vaccine was able to inhibit virus transmission from the host to the vector. IMPORTANCE Zika is a mosquito-borne viral disease, causing acute debilitating symptoms and complications in infected individuals and irreversible neuronal abnormalities in newborn children. The primary vectors of ZIKV are Aedes aegypti mosquitoes. Despite representing a significant public health burden with a widespread transmission in many regions of the world, Zika remains a neglected disease with no effective antiviral therapies or approved vaccines. It is known that components of the mosquito bite lead to an enhancement of viral infection and spread, but this aspect is often overlooked when vaccine candidates undergo preclinical validation. In this study, we included mosquitoes as viral vectors, demonstrating the ability of a promising vaccine candidate to protect animals against ZIKV infections after the bite of an infected mosquito and to also prevent its further transmission. These findings represent an additional crucial step for the development of an effective prevention tool for clinical use.
Collapse
Affiliation(s)
| | - Rapeepat Tandavanitj
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Biologicals Research Group, Research and Development Institute, Government Pharmaceutical Organization, Bangkok, Thailand
| | - Thomas H. Ant
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Shivan M. Murdochy
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Daniel D. Gingell
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Chayanee Setthapramote
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Piyatida Natsrita
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Alain Kohl
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Steven P. Sinkins
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Arvind H. Patel
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Giuditta De Lorenzo
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| |
Collapse
|
15
|
Wang K, Zou S, Chen H, Higazy D, Gao X, Zhang Y, Cao S, Cui M. Zika virus replication on endothelial cells and invasion into the central nervous system by inhibiting interferon β translation. Virology 2023; 582:23-34. [PMID: 36996689 DOI: 10.1016/j.virol.2023.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
The blood-brain barrier (BBB) is one of the tightest physical barriers to prevent pathogens from invading the central nervous system (CNS). However, the mechanism by which Zika virus (ZIKV) crossing the BBB remains unresolved. We found ZIKV induced high morbidity and mortality in newborn mice, accompanied by inflammatory injury on CNS. ZIKV was found to replicate primarily in the cortex and hippocampus in neonatal mouse brains. An in vitro model revealed that ZIKV had no impact on hBMECs permeability but led to endothelial activation, as shown by the enhancement of adhesion molecules expression and F-actin redistribution. ZIKV replication in hBMECs might be associated with the suppression of IFN-β translation via inhibiting RPS6 phosphorylation. On the other hand, ZIKV infection induced IFN-stimulated genes (ISGs), activated the mitogen-activated protein kinase (MAPK) signaling pathway, and promoted chemokine secretion. This study provides an understanding of virus replication and transmigration across the BBB during ZIKV infection.
Collapse
|
16
|
Yeh SC, Strilets T, Tan WL, Castillo D, Medkour H, Rey-Cadilhac F, Serrato-Pomar IM, Rachenne F, Chowdhury A, Chuo V, Azar SR, Singh MK, Hamel R, Missé D, Kini RM, Kenney LJ, Vasilakis N, Marti-Renom MA, Nir G, Pompon J, Garcia-Blanco MA. The anti-immune dengue subgenomic flaviviral RNA is present in vesicles in mosquito saliva and is associated with increased infectivity. PLoS Pathog 2023; 19:e1011224. [PMID: 36996041 PMCID: PMC10062553 DOI: 10.1371/journal.ppat.1011224] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023] Open
Abstract
Mosquito transmission of dengue viruses to humans starts with infection of skin resident cells at the biting site. There is great interest in identifying transmission-enhancing factors in mosquito saliva in order to counteract them. Here we report the discovery of high levels of the anti-immune subgenomic flaviviral RNA (sfRNA) in dengue virus 2-infected mosquito saliva. We established that sfRNA is present in saliva using three different methods: northern blot, RT-qPCR and RNA sequencing. We next show that salivary sfRNA is protected in detergent-sensitive compartments, likely extracellular vesicles. In support of this hypothesis, we visualized viral RNAs in vesicles in mosquito saliva and noted a marked enrichment of signal from 3'UTR sequences, which is consistent with the presence of sfRNA. Furthermore, we show that incubation with mosquito saliva containing higher sfRNA levels results in higher virus infectivity in a human hepatoma cell line and human primary dermal fibroblasts. Transfection of 3'UTR RNA prior to DENV2 infection inhibited type I and III interferon induction and signaling, and enhanced viral replication. Therefore, we posit that sfRNA present in salivary extracellular vesicles is delivered to cells at the biting site to inhibit innate immunity and enhance dengue virus transmission.
Collapse
Affiliation(s)
- Shih-Chia Yeh
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Tania Strilets
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Wei-Lian Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - David Castillo
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Hacène Medkour
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | | | | | | | - Avisha Chowdhury
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Vanessa Chuo
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Sasha R. Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Moirangthem Kiran Singh
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Rodolphe Hamel
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - R. Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Linda J. Kenney
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, University of University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Vector-Borne and Zoonotic Diseases, University of University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Preventive Medicine and Population Health, University of University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center for Emerging Viruses and Arboviruses, University of University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marc A. Marti-Renom
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Guy Nir
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Julien Pompon
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - Mariano A. Garcia-Blanco
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
17
|
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
|
18
|
Visser I, Koenraadt CJ, Koopmans MP, Rockx B. The significance of mosquito saliva in arbovirus transmission and pathogenesis in the vertebrate host. One Health 2023. [DOI: 10.1016/j.onehlt.2023.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
|
19
|
Shang YF, Shen YY, Zhang MC, Lv MC, Wang TY, Chen XQ, Lin J. Progress in salivary glands: Endocrine glands with immune functions. Front Endocrinol (Lausanne) 2023; 14:1061235. [PMID: 36817607 PMCID: PMC9935576 DOI: 10.3389/fendo.2023.1061235] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
The production and secretion of saliva is an essential function of the salivary glands. Saliva is a complicated liquid with different functions, including moistening, digestion, mineralization, lubrication, and mucosal protection. This review focuses on the mechanism and neural regulation of salivary secretion, and saliva is secreted in response to various stimuli, including odor, taste, vision, and mastication. The chemical and physical properties of saliva change dynamically during physiological and pathophysiological processes. Moreover, the central nervous system modulates salivary secretion and function via various neurotransmitters and neuroreceptors. Smell, vision, and taste have been investigated for the connection between salivation and brain function. The immune and endocrine functions of the salivary glands have been explored recently. Salivary glands play an essential role in innate and adaptive immunity and protection. Various immune cells such as B cells, T cells, macrophages, and dendritic cells, as well as immunoglobins like IgA and IgG have been found in salivary glands. Evidence supports the synthesis of corticosterone, testosterone, and melatonin in salivary glands. Saliva contains many potential biomarkers derived from epithelial cells, gingival crevicular fluid, and serum. High level of matrix metalloproteinases and cytokines are potential markers for oral carcinoma, infectious disease in the oral cavity, and systemic disease. Further research is required to monitor and predict potential salivary biomarkers for health and disease in clinical practice and precision medicine.
Collapse
Affiliation(s)
- Yu Feng Shang
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
| | - Yi Yang Shen
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
| | - Meng Chen Zhang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Min Chao Lv
- Department of Orthopedics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Tong Ying Wang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Xue Qun Chen
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Department of Neurobiology, Department of Neurology of the Second Affiliated Hospital, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Jun Lin
- Department of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang University School of Stomatology, Hangzhou, China
- *Correspondence: Jun Lin,
| |
Collapse
|
20
|
Jin L, Jiang BG, Yin Y, Guo J, Jiang JF, Qi X, Crispell G, Karim S, Cao WC, Lai R. Interference with LTβR signaling by tick saliva facilitates transmission of Lyme disease spirochetes. Proc Natl Acad Sci U S A 2022; 119:e2208274119. [PMID: 36383602 PMCID: PMC9704693 DOI: 10.1073/pnas.2208274119] [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: 05/13/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Lyme spirochetes have coevolved with ticks to optimize transmission to hosts using tick salivary molecules (TSMs) to counteract host defenses. TSMs modulate various molecular events at the tick-host interface. Lymphotoxin-beta receptor (LTβR) is a vital immune receptor and plays protective roles in host immunity against microbial infections. We found that Ltbr knockout mice were more susceptible to Lyme disease spirochetes, suggesting the involvement of LTβR signaling in tick-borne Borrelia infection. Further investigation showed that a 15-kDa TSM protein from Ixodes persulcatus (I. persulcatus salivary protein; IpSAP) functioned as an immunosuppressant to facilitate the transmission and infection of Lyme disease spirochetes. IpSAP directly interacts with LTβR to block its activation, thus inhibiting the downstream signaling and consequently suppressing immunity. IpSAP immunization provided mice with significant protection against I. persulcatus-mediated Borrelia garinii infection. Notably, the immunization showed considerable cross-protection against other Borrelia infections mediated by other ixodid ticks. One of the IpSAP homologs from other ixodid ticks showed similar effects on Lyme spirochete transmission. Together, our findings suggest that LTβR signaling plays an important role in blocking the transmission and pathogenesis of tick-borne Lyme disease spirochetes, and that IpSAP and its homologs are promising candidates for broad-spectrum vaccine development.
Collapse
Affiliation(s)
- Lin Jin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100101, China
| | - Yizhu Yin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - Jingya Guo
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100101, China
| | - Xiaopeng Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - Gary Crispell
- Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406
| | - Shahid Karim
- Cell and Molecular Biology, School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100101, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| |
Collapse
|
21
|
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: 12] [Impact Index Per Article: 6.0] [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
|
22
|
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: 1.0] [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
|
23
|
Wang Y, Ling L, Zhang Z, Marin-Lopez A. Current Advances in Zika Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10111816. [PMID: 36366325 PMCID: PMC9694033 DOI: 10.3390/vaccines10111816] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV), an emerging arthropod-borne flavivirus, was first isolated in Uganda in 1947 from monkeys and first detected in humans in Nigeria in 1952; it has been associated with a dramatic burden worldwide. Since then, interventions to reduce the burden of ZIKV infection have been mainly restricted to mosquito control, which in the end proved to be insufficient by itself. Hence, the situation prompted scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elucidated. Understanding the viral disease mechanism will provide a better landscape to develop prophylactic and therapeutic strategies against ZIKV. Currently, no specific vaccines or drugs have been approved for ZIKV. However, some are undergoing clinical trials. Notably, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, viral vectors, virus-like particles (VLPs), inactivated virus, live attenuated virus, peptide and protein-based vaccines, passive immunizations by using monoclonal antibodies (MAbs), and vaccines that target vector-derived antigens. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA titers, both in vitro and in vivo. This review provides a comprehensive summary of current advancements in the development of vaccines against Zika virus.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
- Correspondence:
| | - Lin Ling
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Zilei Zhang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
24
|
Identification of Aedes aegypti salivary gland proteins interacting with human immune receptor proteins. PLoS Negl Trop Dis 2022; 16:e0010743. [PMID: 36070318 PMCID: PMC9484696 DOI: 10.1371/journal.pntd.0010743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/19/2022] [Accepted: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Mosquito saliva proteins modulate the human immune and hemostatic systems and control mosquito-borne pathogenic infections. One mechanism through which mosquito proteins may influence host immunity and hemostasis is their interactions with key human receptor proteins that may act as receptors for or coordinate attacks against invading pathogens. Here, using pull-down assays and proteomics-based mass spectrometry, we identified 11 Ae. aegypti salivary gland proteins (SGPs) (e.g., apyrase, Ae. aegypti venom allergen-1 [AaVA-1], neutrophil stimulating protein 1 [NeSt1], and D7 proteins), that interact with one or more of five human receptor proteins (cluster of differentiation 4 [CD4], CD14, CD86, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin [DC-SIGN], and Toll-like receptor 4 [TLR4]). We focused on CD4- and DC-SIGN-interacting proteins and confirmed that CD4 directly interacts with AaVA-1, D7, and NeST1 recombinant proteins and that AaVA-1 showed a moderate interaction with DC-SIGN using ELISA. Bacteria responsive protein 1 (AgBR1), an Ae. aegypti saliva protein reported to enhance ZIKV infection in humans but that was not identified in our pull-down assay moderately interacts with CD4 in the ELISA assay. Functionally, we showed that AaVA-1 and NeST1 proteins promoted activation of CD4+ T cells. We propose the possible impact of these interactions and effects on mosquito-borne viral infections such as dengue, Zika, and chikungunya viruses. Overall, this study provides key insight into the vector-host (protein-protein) interaction network and suggests roles for these interactions in mosquito-borne viral infections. Here, we report our results from a pull-down assay and ELISA, which identified Ae. aegypti salivary gland proteins that interact with one or more of five human receptor proteins. Some of these interactions could affect the expression of costimulatory molecules involved in host defense against pathogens. This underscores the potential proviral or antiviral roles of these interactions on mosquito-borne viral infections. Our study provides a preliminary enquiry into the vector (mosquito)-host (human) interaction networks and how this interaction could be further investigated and harnessed as a strategy to augment existing vector-borne diseases control approaches.
Collapse
|
25
|
Mosquito saliva enhances virus infection through sialokinin-dependent vascular leakage. Proc Natl Acad Sci U S A 2022; 119:e2114309119. [PMID: 35675424 PMCID: PMC9214539 DOI: 10.1073/pnas.2114309119] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Viruses transmitted by Aedes mosquitoes are an increasingly important global cause of disease. Defining common determinants of host susceptibility to this large group of heterogenous pathogens is key for informing the rational design of panviral medicines. Infection of the vertebrate host with these viruses is enhanced by mosquito saliva, a complex mixture of salivary-gland-derived factors and microbiota. We show that the enhancement of infection by saliva was dependent on vascular function and was independent of most antisaliva immune responses, including salivary microbiota. Instead, the Aedes gene product sialokinin mediated the enhancement of virus infection through a rapid reduction in endothelial barrier integrity. Sialokinin is unique within the insect world as having a vertebrate-like tachykinin sequence and is absent from Anopheles mosquitoes, which are incompetent for most arthropod-borne viruses, whose saliva was not proviral and did not induce similar vascular permeability. Therapeutic strategies targeting sialokinin have the potential to limit disease severity following infection with Aedes-mosquito-borne viruses.
Collapse
|
26
|
Duan Z, Zhang J, Chen X, Liu M, Zhao H, Jin L, Zhang Z, Luan N, Meng P, Wang J, Tan Z, Li Y, Deng G, Lai R. Role of LL-37 in thrombotic complications in patients with COVID-19. Cell Mol Life Sci 2022; 79:309. [PMID: 35596804 PMCID: PMC9123294 DOI: 10.1007/s00018-022-04309-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/28/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Blood clot formation induced by dysfunctional coagulation is a frequent complication of coronavirus disease 2019 (COVID-19) and a high-risk factor for severe illness and death. Neutrophil extracellular traps (NETs) are implicated in COVID-19-induced immunothrombosis. Furthermore, human cathelicidin, a NET component, can perturb the interaction between the SARS-CoV-2 spike protein and its ACE2 receptor, which mediates viral entry into cells. At present, however, the levels of cathelicidin antimicrobial peptides after SARS-CoV-2 infection and their role in COVID-19 thrombosis formation remain unclear. In the current study, we analyzed coagulation function and found a decrease in thrombin time but an increase in fibrinogen level, prothrombin time, and activated partial thromboplastin time in COVID-19 patients. In addition, the cathelicidin antimicrobial peptide LL-37 was upregulated by the spike protein and significantly elevated in the plasma of patients. Furthermore, LL-37 levels were negatively correlated with thrombin time but positively correlated with fibrinogen level. In addition to platelet activation, cathelicidin peptides enhanced the activity of coagulation factors, such as factor Xa (FXa) and thrombin, which may induce hypercoagulation in diseases with high cathelicidin peptide levels. Injection of cathelicidin peptides promoted the formation of thrombosis, whereas deletion of cathelicidin inhibited thrombosis in vivo. These results suggest that cathelicidin antimicrobial peptide LL-37 is elevated during SARS-CoV-2 infection, which may induce hypercoagulation in COVID-19 patients by activating coagulation factors.
Collapse
Affiliation(s)
- Zilei Duan
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Juan Zhang
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Xue Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ming Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Hongwen Zhao
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Lin Jin
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Zhiye Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ning Luan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China
| | - Ping Meng
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650041, Yunnan, China
| | - Jing Wang
- Department of Laboratory Diagnosis, Chongqing Public Health Medical Center, Public Health Hospital of Southwest University, 109 Baoyu Rd. Shapingba, Chongqing, 400038, China
| | - Zhaoxia Tan
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Yaxiong Li
- Department of Cardiovascular Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, 650041, Yunnan, China.
| | - Guohong Deng
- Southwest Hospital, Third Military Medical University (Army Medical University, 29 Gaotanyan Street, Shapingba, Chongqing, 400038, China.
| | - Ren Lai
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, 511458, China.
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Sino-African Joint Research Center, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China.
| |
Collapse
|
27
|
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
|
28
|
A leafhopper saliva protein mediates horizontal transmission of viral pathogens from insect vectors into rice phloem. Commun Biol 2022; 5:204. [PMID: 35246603 PMCID: PMC8897447 DOI: 10.1038/s42003-022-03160-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022] Open
Abstract
Numerous insects transmit viruses together with saliva to plant phloem, but the roles of saliva components remain elusive. Here, we report that calcium-binding protein (CBP), a universal insect saliva protein, is modified to benefit horizontal transmission of a devastating rice reovirus into plant phloem. CBP effectively competes with virus-induced filaments to target and traverse actin-based apical plasmalemma into saliva-stored cavities in salivary glands of leafhopper vector. Thus, the inhibition of CBP expression by viral infection facilitates filament-mediated viral secretion into salivary cavities and then into plant phloem. Furthermore, virus-mediated reduction of CBP secretion causes an increase of cytosolic Ca2+ levels in rice, triggering substantial callose deposition and H2O2 production. Thus, viruliferous vectors encounter stronger feeding barriers, probe more frequently, and secrete more saliva into plants, ultimately enhancing viral transmission. We thus conclude that the inhibition of CBP secretion facilitates viral secretion and increases host defense response to benefit viral transmission. CBP, a calcium binding protein found in insect saliva, allows for the transmission of the devastating rice gall dwarf virus into plant phloem. This interaction with CBP is compounded by stronger feeding barriers, more frequent probing behavior, and increased saliva secretion into plants by insect vectors, all increasing the likelihood of viral transmission.
Collapse
|
29
|
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: 4.0] [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
|
30
|
Zhu T, Chen X, Qiu H, Liu Y, Mwangi J, Zhao L, Ding W, Lai R, Jin L. Aspirin Alleviates Particulate Matter Induced Asymptomatic Orchitis of Mice via Suppression of cGAS-STING Signaling. Front Immunol 2021; 12:734546. [PMID: 34925318 PMCID: PMC8673441 DOI: 10.3389/fimmu.2021.734546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
As an important source of air pollutant, airborne particulate matter (PM) has become a major threat to public health. Orchitis is characterized by acute or chronic testicular inflammation and is a primary cause of male infertility. Although accumulating evidence indicates that PM exposure is associated with increased male infertility rates, the mechanism by which PM is involved is not well understood. Here, we found that short-term PM exposure activated NF-κB signaling in mouse Leydig cells and testes and leading to asymptomatic orchitis. Analyzing the mitochondrial abundance and cGAMP levels in PM exposed mouse Leydig cells, we found that PM exposure induced mitochondrial injury and mtDNA release, leading to inflammation via the cGAS-STING axis. We also found that aspirin-induced acetylation of cGAS inhibited the inflammation in mice after PM exposure, especially in the testes. Moreover, aspirin pretreatment rescued offspring growth in PM-exposed mice. In summary, our study not only provides evidence that PM-induced asymptomatic orchitis in mice may be amenable to aspirin pre-treatment by acetylating cGAS, but also provides a potential explanation for male infertility caused by air pollutants.
Collapse
Affiliation(s)
- Tengyu Zhu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China.,College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Xue Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Huan Qiu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Yang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - James Mwangi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Ling Zhao
- Experimental Animal Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenjun Ding
- Laboratory of Environment and Health, University of Chinese Academy of Sciences, Beijing, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China.,Kunming institute of zoology-the Chinese university of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Lin Jin
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
31
|
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
|
32
|
Manning JE, Chea S, Parker DM, Bohl JA, Lay S, Mateja A, Man S, Nhek S, Ponce A, Sreng S, Kong D, Kimsan S, Meneses C, Fay MP, Suon S, Huy R, Lon C, Leang R, Oliveira F. Development of inapparent dengue associated with increased antibody levels to Aedes aegypti salivary proteins: a longitudinal dengue cohort in Cambodia. J Infect Dis 2021; 226:1327-1337. [PMID: 34718636 DOI: 10.1093/infdis/jiab541] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/26/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We established the first prospective cohort to understand how infection with dengue virus is influenced by vector-specific determinants like humoral immunity to Aedes aegypti salivary proteins. METHODS Children aged two to nine years old enrolled in the PAGODAS (Pediatric Assessment Group of Dengue and Aedes Saliva) cohort with informed consent by their guardians. Children were followed semi-annually for antibodies to dengue and to proteins in Ae. aegypti salivary gland homogenate using enzyme-linked immunosorbent assays and dengue-specific neutralization titers. Children presented with fever at any time for dengue testing. RESULTS From July 13 to August 30, 2018, we enrolled 771 children. At baseline, 22% (173/770) had evidence of neutralizing antibodies to one or more dengue serotypes. By April 2020, 51 children had symptomatic dengue while 148 dengue-naïve children had inapparent dengue defined by neutralization assays. In a multivariate model, individuals with higher antibodies to Ae. aegypti salivary proteins were 1.5x more likely to have dengue infection (HR 1.47 95% CI 1.05-2.06; p=0.02), particularly individuals with inapparent dengue (HR 1.64 95% CI 1.12-2.41; p=0.01). CONCLUSIONS High levels of seropositivity to Ae. aegypti salivary proteins are associated with future development of dengue infection, primarily inapparent, in dengue-naïve Cambodian children.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,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.,National Center for Parasitology, Entomology, and Malaria Control, Ministry 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, Bethesda, Maryland, USA
| | - Sreyngim Lay
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Somnang Man
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Sreynik Nhek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Aiyana Ponce
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sokunthea Sreng
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Dara Kong
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Soun Kimsan
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia.,National Dengue Control Program, Ministry of Health, Phnom Penh, Cambodia
| | - Claudio Meneses
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael P Fay
- Biostatistics Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Seila Suon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.,National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia
| | - Chanthap Lon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh Cambodia.,National Dengue Control Program, Ministry of Health, Phnom Penh, Cambodia
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Schneider CA, Calvo E, Peterson KE. Arboviruses: How Saliva Impacts the Journey from Vector to Host. Int J Mol Sci 2021; 22:ijms22179173. [PMID: 34502092 PMCID: PMC8431069 DOI: 10.3390/ijms22179173] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 12/21/2022] Open
Abstract
Arthropod-borne viruses, referred to collectively as arboviruses, infect millions of people worldwide each year and have the potential to cause severe disease. They are predominately transmitted to humans through blood-feeding behavior of three main groups of biting arthropods: ticks, mosquitoes, and sandflies. The pathogens harbored by these blood-feeding arthropods (BFA) are transferred to animal hosts through deposition of virus-rich saliva into the skin. Sometimes these infections become systemic and can lead to neuro-invasion and life-threatening viral encephalitis. Factors intrinsic to the arboviral vectors can greatly influence the pathogenicity and virulence of infections, with mounting evidence that BFA saliva and salivary proteins can shift the trajectory of viral infection in the host. This review provides an overview of arbovirus infection and ways in which vectors influence viral pathogenesis. In particular, we focus on how saliva and salivary gland extracts from the three dominant arbovirus vectors impact the trajectory of the cellular immune response to arbovirus infection in the skin.
Collapse
Affiliation(s)
- Christine A. Schneider
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA;
| | - Karin E. Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
- Correspondence:
| |
Collapse
|
34
|
Liu M, Wang G, Xu R, Shen C, Ni H, Lai R. Soy Isoflavones Inhibit Both GPIb-IX Signaling and αIIbβ3 Outside-In Signaling via 14-3-3ζ in Platelet. Molecules 2021; 26:4911. [PMID: 34443497 PMCID: PMC8399232 DOI: 10.3390/molecules26164911] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/30/2022] Open
Abstract
Soy diet is thought to help prevent cardiovascular diseases in humans. Isoflavone, which is abundant in soybean and other legumes, has been reported to possess antiplatelet activity and potential antithrombotic effect. Our study aims to elucidate the potential target of soy isoflavone in platelet. The anti-thrombosis formation effect of genistein and daidzein was evaluated in ex vivo perfusion chamber model under low (300 s-1) and high (1800 s-1) shear forces. The effect of genistein and daidzein on platelet aggregation and spreading was evaluated with platelets from both wildtype and GPIbα deficient mice. The interaction of these soy isoflavone with 14-3-3ζ was detected by surface plasmon resonance (SPR) and co-immunoprecipitation, and the effect of αIIbβ3-mediated outside-in signaling transduction was evaluated by western blot. We found both genistein and daidzein showed inhibitory effect on thrombosis formation in perfusion chamber, especially under high shear force (1800 s-1). These soy isoflavone interact with 14-3-3ζ and inhibited both GPIb-IX and αIIbβ3-mediated platelet aggregation, integrin-mediated platelet spreading and outside-in signaling transduction. Our findings indicate that 14-3-3ζ is a novel target of genistein and daidzein. 14-3-3ζ, an adaptor protein that regulates both GPIb-IX and αIIbβ3-mediated platelet activation is involved in soy isoflavone mediated platelet inhibition.
Collapse
Affiliation(s)
- Ming Liu
- Department of Molecular and Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China;
| | - Gan Wang
- Key Laboratory of Bioactive Peptides, Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650032, China; (G.W.); (R.X.)
| | - Runjia Xu
- Key Laboratory of Bioactive Peptides, Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650032, China; (G.W.); (R.X.)
| | - Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada; (C.S.); (H.N.)
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada; (C.S.); (H.N.)
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON M5G 2M1, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Ren Lai
- Key Laboratory of Bioactive Peptides, Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650032, China; (G.W.); (R.X.)
| |
Collapse
|
35
|
Lara PG, Esteves E, Sales-Campos H, Assis JB, Henrique MO, Barros MS, Neto LS, Silva PI, Martins JO, Cardoso CRB, Ribeiro JMC, Sá-Nunes A. AeMOPE-1, a Novel Salivary Peptide From Aedes aegypti, Selectively Modulates Activation of Murine Macrophages and Ameliorates Experimental Colitis. Front Immunol 2021; 12:681671. [PMID: 34349757 PMCID: PMC8327214 DOI: 10.3389/fimmu.2021.681671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/25/2021] [Indexed: 11/17/2022] Open
Abstract
The sialotranscriptomes of Aedes aegypti revealed a transcript overexpressed in female salivary glands that codes a mature 7.8 kDa peptide. The peptide, specific to the Aedes genus, has a unique sequence, presents a putative secretory nature and its function is unknown. Here, we confirmed that the peptide is highly expressed in the salivary glands of female mosquitoes when compared to the salivary glands of males, and its secretion in mosquito saliva is able to sensitize the vertebrate host by inducing the production of specific antibodies. The synthetic version of the peptide downmodulated nitric oxide production by activated peritoneal murine macrophages. The fractionation of a Ae. aegypti salivary preparation revealed that the fractions containing the naturally secreted peptide reproduced the nitric oxide downmodulation. The synthetic peptide also selectively interfered with cytokine production by murine macrophages, inhibiting the production of IL-6, IL-12p40 and CCL2 without affecting TNF-α or IL-10 production. Likewise, intracellular proteins associated with macrophage activation were also distinctively modulated: while iNOS and NF-κB p65 expression were diminished, IκBα and p38 MAPK expression did not change in the presence of the peptide. The anti-inflammatory properties of the synthetic peptide were tested in vivo on a dextran sulfate sodium-induced colitis model. The therapeutic administration of the Ae. aegypti peptide reduced the leukocytosis, macrophage activity and nitric oxide levels in the gut, as well as the expression of cytokines associated with the disease, resulting in amelioration of its clinical signs. Given its biological properties in vitro and in vivo, the molecule was termed Aedes-specific MOdulatory PEptide (AeMOPE-1). Thus, AeMOPE-1 is a novel mosquito-derived immunobiologic with potential to treat immune-mediated disorders.
Collapse
Affiliation(s)
- Priscila G. Lara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Eliane Esteves
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Helioswilton Sales-Campos
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Josiane B. Assis
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Maressa O. Henrique
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Michele S. Barros
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Leila S. Neto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Pedro I. Silva
- Laboratory for Applied Toxinology, Butantan Institute, Sao Paulo, Brazil
| | - Joilson O. Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cristina R. B. Cardoso
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - José M. C. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Anderson Sá-Nunes
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- National Institute of Science and Technology in Molecular Entomology, National Council of Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Lymphotoxin β Receptor: a Crucial Role in Innate and Adaptive Immune Responses against Toxoplasma gondii. Infect Immun 2021; 89:IAI.00026-21. [PMID: 33753412 PMCID: PMC8316152 DOI: 10.1128/iai.00026-21] [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: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/21/2022] Open
Abstract
The lymphotoxin β receptor (LTβR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTβR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTβR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTβR-deficient (LTβR−/−) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTβR−/− mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTβR in cytokine regulation and adaptive immune responses to control T. gondii.
Collapse
|
37
|
Olajiga O, Holguin-Rocha AF, Rippee-Brooks M, Eppler M, Harris SL, Londono-Renteria B. Vertebrate Responses against Arthropod Salivary Proteins and Their Therapeutic Potential. Vaccines (Basel) 2021; 9:347. [PMID: 33916367 PMCID: PMC8066741 DOI: 10.3390/vaccines9040347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 01/11/2023] Open
Abstract
The saliva of hematophagous arthropods contains a group of active proteins to counteract host responses against injury and to facilitate the success of a bloodmeal. These salivary proteins have significant impacts on modulating pathogen transmission, immunogenicity expression, the establishment of infection, and even disease severity. Recent studies have shown that several salivary proteins are immunogenic and antibodies against them may block infection, thereby suggesting potential vaccine candidates. Here, we discuss the most relevant salivary proteins currently studied for their therapeutic potential as vaccine candidates or to control the transmission of human vector-borne pathogens and immune responses against different arthropod salivary proteins.
Collapse
Affiliation(s)
- Olayinka Olajiga
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Andrés F. Holguin-Rocha
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | | | - Megan Eppler
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Shanice L. Harris
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| | - Berlin Londono-Renteria
- Vector Biology Laboratory, Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (O.O.); (A.F.H.-R.); (M.E.); (S.L.H.)
| |
Collapse
|
38
|
The 14-3-3ζ-c-Src-integrin-β3 complex is vital for platelet activation. Blood 2021; 136:974-988. [PMID: 32584951 DOI: 10.1182/blood.2019002314] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Several adaptor molecules bind to cytoplasmic tails of β-integrins and facilitate bidirectional signaling, which is critical in thrombosis and hemostasis. Interfering with integrin-adaptor interactions spatially or temporally to inhibit thrombosis without affecting hemostasis is an attractive strategy for the development of safe antithrombotic drugs. We show for the first time that the 14-3-3ζ-c-Src-integrin-β3 complex is formed during platelet activation. 14-3-3ζ-c-Src interaction is mediated by the -PIRLGLALNFSVFYYE- fragment (PE16) on the 14-3-3ζ and SH2-domain on c-Src, whereas the 14-3-3ζ-integrin-β3 interaction is mediated by the -ESKVFYLKMKGDYYRYL- fragment (EL17) on the 14-3-3ζ and -KEATSTF- fragment (KF7) on the β3-integrin cytoplasmic tail. The EL17-motif inhibitor, or KF7 peptide, interferes with the formation of the 14-3-3ζ-c-Src-integrin-β3 complex and selectively inhibits β3 outside-in signaling without affecting the integrin-fibrinogen interaction, which suppresses thrombosis without causing significant bleeding. This study characterized a previously unidentified 14-3-3ζ-c-Src-integrin-β3 complex in platelets and provided a novel strategy for the development of safe and effective antithrombotic treatments.
Collapse
|
39
|
Hameed M, Wahaab A, Nawaz M, Khan S, Nazir J, Liu K, Wei J, Ma Z. Potential Role of Birds in Japanese Encephalitis Virus Zoonotic Transmission and Genotype Shift. Viruses 2021; 13:357. [PMID: 33668224 PMCID: PMC7996159 DOI: 10.3390/v13030357] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/13/2022] Open
Abstract
Japanese encephalitis (JE) is a vaccine-preventable disease caused by the Japanese encephalitis virus (JEV), which is primarily prevalent in Asia. JEV is a Flavivirus, classified into a single serotype with five genetically distinct genotypes (I, II, III, IV, and V). JEV genotype III (GIII) had been the most dominant strain and caused numerous outbreaks in the JEV endemic countries until 1990. However, recent data shows the emergence of JEV genotype I (GI) as a dominant genotype and it is gradually displacing GIII. The exact mechanism of this genotype displacement is still unclear. The virus can replicate in mosquito vectors and vertebrate hosts to maintain its zoonotic life cycle; pigs and aquatic wading birds act as an amplifying/reservoir hosts, and the humans and equines are dead-end hosts. The important role of pigs as an amplifying host for the JEV is well known. However, the influence of other domestic animals, especially birds, that live in high abundance and close proximity to the human is not well studied. Here, we strive to briefly highlight the role of birds in the JEV zoonotic transmission, discovery of birds as a natural reservoirs and amplifying host for JEV, species of birds susceptible to the JEV infection, and the proposed effect of JEV on the poultry industry in the future, a perspective that has been neglected for a long time. We also discuss the recent in vitro and in vivo studies that show that the newly emerged GI viruses replicated more efficiently in bird-derived cells and ducklings/chicks than GIII, and an important role of birds in the JEV genotype shift from GIII to GI.
Collapse
Affiliation(s)
- Muddassar Hameed
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Abdul Wahaab
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Mohsin Nawaz
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Sawar Khan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Jawad Nazir
- Vaccinologist/Head Virology, Tréidlia Biovet Pty Ltd. Units, Seven Hills, NSW 2147, Australia;
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (A.W.); (M.N.); (S.K.); (K.L.)
| |
Collapse
|
40
|
Marin-Lopez A, Wang Y, Jiang J, Ledizet M, Fikrig E. AgBR1 and NeSt1 antisera protect mice from Aedes aegypti-borne Zika infection. Vaccine 2021; 39:1675-1679. [PMID: 33622591 DOI: 10.1016/j.vaccine.2021.01.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/26/2021] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
Zika virus(ZIKV) is primarily spread by Aedes. aegyptimosquitoes. Infection with ZIKV can result in diverse clinical symptoms in humans, ranging from mild to severe. Previously, we demonstrated that passive immunization against A. aegypti AgBR1 or NeSt1 antiserum, two mosquito saliva proteins that are transmitted with the virus, conferred partial protection against ZIKV in mice. Each individual antiserum altered the early host response in the skin and reduced viremia. Here, we show that passive immunization with a combination of AgBR1- and NeSt1-specific antibodies enhanced survival and reduced the viral burden in blood, thereby protecting mice from mosquito-borne ZIKV infection. This finding suggests that targeting a combination of mosquito saliva proteins, with AgBR1 and NeSt1 as model antigens, may be used as a vaccine strategy to help prevent mosquito-borne ZIKV infection.
Collapse
Affiliation(s)
- Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Yuchen Wang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, Hubei 430072, China.
| | - Junjun Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | | | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
41
|
Assis JB, Cogliati B, Esteves E, Capurro ML, Fonseca DM, Sá-Nunes A. Aedes aegypti mosquito saliva ameliorates acetaminophen-induced liver injury in mice. PLoS One 2021; 16:e0245788. [PMID: 33556084 PMCID: PMC7869984 DOI: 10.1371/journal.pone.0245788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) overdose is the most common cause of drug-induced liver injury (DILI). Although the primary hepatic damage is induced by APAP-derived toxic intermediates resulting from cytochrome P450 metabolism, immune components also play an important role in DILI pathophysiology. Aedes aegypti saliva is a source of bioactive molecules with in vitro anti-inflammatory and immunomodulatory activities. However, evidences on the therapeutic use of Ae. aegypti salivary preparations in animal models of relevant clinical conditions are still scarce. Thus, the present study was designed to evaluate the protective role of Ae. aegypti saliva in a murine model of APAP-induced DILI. C57BL/6 mice were exposed to Ae. aegypti bites 2 hours after APAP overdose. Biochemical and immunological parameters were evaluated in blood and liver samples at different time points after APAP administration. Exposure to Ae. aegypti saliva attenuated liver damage, as demonstrated by reduced hepatic necrosis and serum levels of alanine aminotransferase in APAP-overdosed mice. The levels of hepatic CYP2E1, the major enzyme responsible for the bioactivation of APAP, were not changed in Ae. aegypti exposed animals, suggesting no effects in the generation of hepatotoxic metabolites. On the other hand, mice treated with Ae. aegypti saliva following APAP overdose presented lower serum concentration of TNF-α, IL-6, IL-1β and IL-10, as well as reduced frequency of inflammatory cell populations in the liver, such as NKT cells, macrophages and dendritic cells. These findings show that Ae. aegypti saliva has bioactive molecules with therapeutic properties and may represent a prospective source of new compounds in the management of DILI-associated inflammatory disorders and, perhaps, many other inflammatory/autoimmune diseases.
Collapse
Affiliation(s)
- Josiane B. Assis
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruno Cogliati
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Margareth L. Capurro
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise M. Fonseca
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
42
|
Lei V, Petty AJ, Atwater AR, Wolfe SA, MacLeod AS. Skin Viral Infections: Host Antiviral Innate Immunity and Viral Immune Evasion. Front Immunol 2020; 11:593901. [PMID: 33240281 PMCID: PMC7677409 DOI: 10.3389/fimmu.2020.593901] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022] Open
Abstract
The skin is an active immune organ that functions as the first and largest site of defense to the outside environment. Serving as the primary interface between host and pathogen, the skin’s early immune responses to viral invaders often determine the course and severity of infection. We review the current literature pertaining to the mechanisms of cutaneous viral invasion for classical skin-tropic, oncogenic, and vector-borne skin viruses. We discuss the skin’s evolved mechanisms for innate immune viral defense against these invading pathogens, as well as unique strategies utilized by the viruses to escape immune detection. We additionally explore the roles that demographic and environmental factors, such as age, biological sex, and the cutaneous microbiome, play in altering the host immune response to viral threats.
Collapse
Affiliation(s)
- Vivian Lei
- Department of Dermatology, Duke University, Durham, NC, United States.,School of Medicine, Duke University, Durham, NC, United States
| | - Amy J Petty
- School of Medicine, Duke University, Durham, NC, United States
| | - Amber R Atwater
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Sarah A Wolfe
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC, United States.,Department of Immunology, Duke University, Durham, NC, United States.,Pinnell Center for Investigative Dermatology, Duke University, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
43
|
Onyango MG, Ciota AT, Kramer LD. The Vector - Host - Pathogen Interface: The Next Frontier in the Battle Against Mosquito-Borne Viral Diseases? Front Cell Infect Microbiol 2020; 10:564518. [PMID: 33178624 PMCID: PMC7596266 DOI: 10.3389/fcimb.2020.564518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
An unprecedented spread of mosquito-borne viruses and increasing populations of mosquito vectors has led to an increase in the frequency of mosquito-borne virus disease outbreaks. Recent outbreaks of Zika virus (ZIKV) and yellow fever virus (YFV), among others have led to a concerted effort to understand the biology of mosquito-borne viruses and their interaction with their vector mosquito and vertebrate hosts. Recent studies have aimed to understand the vector-host-pathogen interface and how it influences infection, tropism and disease severity in the vertebrate host. The initial replication of the pathogen at the skin bite site is crucial in determining the progression of the infection in the vertebrate host. Delineating the role of the commensal microbes in the mosquito saliva as well as how they interact with the vertebrate host keratinocytes will improve our understanding of disease immunopathology and may lead to new therapeutics.
Collapse
Affiliation(s)
- Maria Gorreti Onyango
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States
| | - Alexander T Ciota
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| | - Laura D Kramer
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| |
Collapse
|
44
|
Guo X, Ma P, Li Y, Yang Y, Wang C, Xu T, Wang H, Li C, Mao B, Qi X. RNF220 mediates K63-linked polyubiquitination of STAT1 and promotes host defense. Cell Death Differ 2020; 28:640-656. [PMID: 32814877 DOI: 10.1038/s41418-020-00609-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 08/02/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
STAT1 is a master regulator that orchestrates type 1 and 2 interferon (IFN)-induced IFN-stimulated gene (ISG) expression. The mechanisms by which STAT1 is phosphorylated and activated upon IFN signaling remain elusive. Our work demonstrated that ubiquitination of STAT1 mediated by the E3 ligase RNF220 contributed significantly to STAT1 activation and innate immune responses. Rnf220 gene deficiency resulted in the downregulation of IFN signaling and decreased expression of ISGs in response to type 1 and 2 IFNs stimulation and Acinetobacter baumannii and HSV-1 infection. Mechanistically, RNF220 interacted with STAT1 and mediated the K63-linked polyubiquitination of STAT1 at residue K110, which promoted the interaction between STAT1 and the kinase JAK1. The expression of RNF220 was induced by pathogenic infection and IFN signaling. RNF220 promoted STAT1 ubiquitination and phosphorylation through a positive feedback loop. RNF220 haploinsufficiency impaired IFN signaling, and RNF220-defective mice were more susceptible to A. baumannii and HSV-1 infection than WT mice. Our work offers novel insights into the mechanisms of STAT1 modulation and provides potential therapeutic targets against bacterial and viral infection and inflammatory diseases.
Collapse
Affiliation(s)
- Xiaomin Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China.,Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
| | - Yuwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Yanan Yang
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Chaoming Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Tao Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
| | - Huishan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, 650204, Kunming, Yunnan, China
| | - Chaocui Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China. .,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, 650223, Kunming, China.
| | - Xiaopeng Qi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China. .,Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China.
| |
Collapse
|
45
|
Guerrero D, Cantaert T, Missé D. Aedes Mosquito Salivary Components and Their Effect on the Immune Response to Arboviruses. Front Cell Infect Microbiol 2020; 10:407. [PMID: 32850501 PMCID: PMC7426362 DOI: 10.3389/fcimb.2020.00407] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Vector-borne diseases are responsible for over a billion infections each year and nearly one million deaths. Mosquito-borne dengue virus, West Nile, Japanese encephalitis, Zika, Chikungunya, and Rift Valley Fever viruses constitute major public health problems in regions with high densities of arthropod vectors. During the initial step of the transmission cycle, vector, host, and virus converge at the bite site, where local immune cells interact with the vector's saliva. Hematophagous mosquito saliva is a mixture of bioactive components known to modulate vertebrate hemostasis, immunity, and inflammation during the insect's feeding process. The capacity of mosquito saliva to modulate the host immune response has been well-studied over the last few decades and has led to the consensus that the presence of saliva is linked to the enhancement of virus transmission, host susceptibility, disease progression, viremia levels, and mortality. We review some of the major aspects of the interactions between mosquito saliva and the host immune response that may be useful for future studies on the control of arboviruses.
Collapse
Affiliation(s)
- David Guerrero
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Dorothée Missé
- MIVEGEC, IRD, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
46
|
Experimental Infection of Newly Hatched Domestic Ducklings via Japanese Encephalitis Virus-Infected Mosquitoes. Pathogens 2020; 9:pathogens9050371. [PMID: 32408553 PMCID: PMC7281460 DOI: 10.3390/pathogens9050371] [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: 04/13/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 11/30/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a zoonotic pathogen that is maintained by mosquito vectors and vertebrate hosts including birds in a natural transmission cycle. Domestic ducklings are sensitive to JEV infection, but the clinical responses of domestic ducklings to natural JEV infection are unknown. In this study, we simulated the natural JEV infection of domestic ducklings via JEV-infected mosquito bites to evaluate the pathogenicity of JEV in domestic ducklings. Specific pathogen-free domestic ducklings were infected at day 2 post-hatching with JEV-infected Culex pipiens mosquito bites and monitored for clinical responses. Among 20 ducklings exposed to JEV-infected mosquitoes, six showed mild and non-characteristic clinical signs starting at two days post-infection, then died suddenly with neurological signs of opisthotonos (a condition of spasm of the back muscles causing the head and limbs to bend backward and the trunk to arch forward) between two and three days post-infection. The mortality of the affected ducklings was 30% (6/20). Multifocal lymphohistiocytic perivascular cuffs and lymphohistiocytic meningitis were macroscopically observed in the affected duckling brains. JEV was detected in the cytoplasm of neuronal cells in the affected duckling brains by immunohistochemical assays and was recovered from the affected duckling brains by viral isolation. These observations indicated that JEV infection via mosquito bites causes mortality associated with viral encephalitis in newly hatched domestic ducklings, thus demonstrating the potential pathogenicity of JEV in domestic ducklings under natural conditions.
Collapse
|
47
|
Autophagy Contributes to Host Immunity and Protection against Zika Virus Infection via Type I IFN Signaling. Mediators Inflamm 2020; 2020:9527147. [PMID: 32410874 PMCID: PMC7204160 DOI: 10.1155/2020/9527147] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/20/2020] [Accepted: 04/08/2020] [Indexed: 11/17/2022] Open
Abstract
Recent studies have indicated that the Zika virus (ZIKV) has a significant impact on the fetal brain, and autophagy is contributing to host immune response and defense against virus infection. Here, we demonstrate that ZIKV infection triggered increased LC3 punctuation in mouse monocyte-macrophage cell line (RAW264.7), mouse microglial cell line (BV2), and hindbrain tissues, proving the occurrence of autophagy both in vitro and in vivo. Interestingly, manual intervention of autophagy, like deficiency inhibited by 3-MA, can reduce viral clearance in RAW264.7 cells upon ZIKV infection. Besides, specific siRNA strategy confirmed that autophagy can be activated through Atg7-Atg5 and type I IFN signaling pathway upon ZIKV infection, while knocking down of Atg7 and Atg5 effectively decreased the ZIKV clearance in phagocytes. Furthermore, we analyzed that type I IFN signaling could contribute to autophagic clearance of invaded ZIKV in phagocytes. Taken together, our findings demonstrate that ZIKV-induced autophagy is favorable to activate host immunity, particularly through type I IFN signaling, which participates in host protection and defense against ZIKV infection.
Collapse
|
48
|
Wang Y, Marin-Lopez A, Jiang J, Ledizet M, Fikrig E. Vaccination with Aedes aegypti AgBR1 Delays Lethal Mosquito-Borne Zika Virus Infection in Mice. Vaccines (Basel) 2020; 8:vaccines8020145. [PMID: 32218189 PMCID: PMC7348886 DOI: 10.3390/vaccines8020145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/10/2020] [Accepted: 03/19/2020] [Indexed: 12/17/2022] Open
Abstract
Zika Virus (ZIKV) is transmitted primarily by Aedes aegypti mosquitoes, resulting in asymptomatic infection, or acute illness with a fever and headache, or neurological complications, such as Guillain-Barre syndrome or fetal microcephaly. Previously, we determined that AgBR1, a mosquito salivary protein, induces inflammatory responses at the bite site, and that passive immunization with AgBR1 antiserum influences mosquito-transmitted ZIKV infection. Here, we show that the active immunization of mice with AgBR1 adjuvanted with aluminum hydroxide delays lethal mosquito-borne ZIKV infection, suggesting that AgBR1 may be used as part of a vaccine to combat ZIKV.
Collapse
Affiliation(s)
- Yuchen Wang
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan 430072, China
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Correspondence: or (Y.W.); (A.M.-L.)
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Correspondence: or (Y.W.); (A.M.-L.)
| | - Junjun Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- School of Public Health, Guangxi Medical University, Nanning 530021, China
| | | | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA; (J.J.); (E.F.)
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
49
|
Giachetto PF, Cunha RC, Nhani A, Garcia MV, Ferro JA, Andreotti R. Gene Expression in the Salivary Gland of Rhipicephalus (Boophilus) microplus Fed on Tick-Susceptible and Tick-Resistant Hosts. Front Cell Infect Microbiol 2020; 9:477. [PMID: 32039052 PMCID: PMC6985549 DOI: 10.3389/fcimb.2019.00477] [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: 10/01/2019] [Accepted: 12/24/2019] [Indexed: 01/10/2023] Open
Abstract
The success of cattle tick fixation largely depends on the secretion of substances that alter the immune response of the host. The majority of these substances are expressed by the parasite salivary gland and secreted in tick saliva. It is known that hosts can mount immune responses against ticks and bovine European breeds, and bovine industrial crossbreeds are more susceptible to infestations than are Bos indicus cattle. To identify candidates for the development of novel control strategies for the cattle tick Rhipicephalus (Boophilus) microplus, a salivary gland transcriptome analysis of engorged females fed on susceptible or resistant hosts was performed. Using RNA-Seq, transcriptomes were de novo assembled and produced a total of 235,451 contigs with 93.3% transcriptome completeness. Differential expression analysis identified 137 sequences as differentially expressed genes (DEGs) between ticks raised on tick-susceptible or tick-resistant cattle. DEGs predicted to be secreted proteins include innexins, which are transmembrane proteins that form gap junction channels; the transporters Na+/dicarboxylate, Na+/tricarboxylate, and phosphate transporter and a putative monocarboxylate transporter; a phosphoinositol 4-phosphate adaptor protein; a cysteine-rich protein containing a trypsin inhibitor-like (TIL) domain; a putative defense protein 3 containing a reeler domain; and an F-actin-uncapping protein LRRC16A with a CARMIL_C domain; these genes were upregulated in ticks fed on tick-susceptible cattle. DEGs predicted to be non-secreted proteins included a small heat shock protein and the negative elongation factor B-like, both acting in a coordinated manner to increase HSP transcript levels in the salivary glands of the ticks fed on tick-susceptible cattle; the 26S protease regulatory subunit 6B and another chaperone with similarity to calnexin, also upregulated in ticks fed on tick-susceptible cattle; an EF-hand calcium binding protein and a serine carboxypeptidase (SCP), both involved in the blood coagulation cascade and upregulated in ticks fed on tick-susceptible cattle; and two ribosomal proteins, the 60S acidic ribosomal protein P2 and the 60S ribosomal protein L19. These results help to characterize cattle tick salivary gland gene expression in tick-susceptible and tick-resistant hosts and suggest new putative targets for the control of tick infestations, as those genes involved in the mechanism of stress response during blood feeding.
Collapse
Affiliation(s)
| | - Rodrigo Casquero Cunha
- Bolsista do CNPq (157460/2018-5), Programa de Pós-Graduação em Biotecnologia, Universidade Federal de Pelotas, Pelotas, Brazil
| | | | | | | | | |
Collapse
|
50
|
Sun P, Nie K, Zhu Y, Liu Y, Wu P, Liu Z, Du S, Fan H, Chen CH, Zhang R, Wang P, Cheng G. A mosquito salivary protein promotes flavivirus transmission by activation of autophagy. Nat Commun 2020; 11:260. [PMID: 31937766 PMCID: PMC6959235 DOI: 10.1038/s41467-019-14115-z] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/07/2019] [Indexed: 01/07/2023] Open
Abstract
Transmission from an infected mosquito to a host is an essential process in the life cycle of mosquito-borne flaviviruses. Numerous studies have demonstrated that mosquito saliva facilitates viral transmission. Here we find that a saliva-specific protein, named Aedes aegypti venom allergen-1 (AaVA-1), promotes dengue and Zika virus transmission by activating autophagy in host immune cells of the monocyte lineage. The AG6 mice (ifnar1–/–ifngr1–/–) bitten by the virus-infected AaVA-1-deficient mosquitoes present a lower viremia and prolonged survival. AaVA-1 intracellularly interacts with a dominant negative binder of Beclin-1, known as leucine-rich pentatricopeptide repeat-containing protein (LRPPRC), and releases Beclin-1 from LRPPRC-mediated sequestration, thereby enabling the initialization of downstream autophagic signaling. A deficiency in Beclin-1 reduces viral infection in mice and abolishes AaVA-1-mediated enhancement of ZIKV transmission by mosquitoes. Our study provides a mechanistic insight into saliva-aided viral transmission and could offer a potential prophylactic target for reducing flavivirus transmission. Mosquito saliva affects transmission of flaviviruses, but underlying mechanisms are incompletely understood. Here, the authors show that Aedes aegypti venom allergen-1 (AaVA-1) promotes dengue and Zika virus transmission by activating autophagy in host immune cells of the monocyte lineage.
Collapse
Affiliation(s)
- Peng Sun
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.,Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China
| | - Kaixiao Nie
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yibin Zhu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.,Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yang Liu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Pa Wu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ziwen Liu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Senyan Du
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Huahao Fan
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli, Taiwan, 35053, China
| | - Renli Zhang
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, The University of Connecticut Health Center, Farmington, Connecticut, 06030, USA
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China. .,Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|