1
|
Khan A, Zakirullah, Wahab S, Hong ST. Advances in antiviral strategies targeting mosquito-borne viruses: cellular, viral, and immune-related approaches. Virol J 2025; 22:26. [PMID: 39905499 PMCID: PMC11792744 DOI: 10.1186/s12985-025-02622-z] [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: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Mosquito-borne viruses (MBVs) are a major global health threat, causing significant morbidity and mortality. MBVs belong to several distinct viral families, each with unique characteristics. The primary families include Flaviviridae (e.g., Dengue, Zika, West Nile, Yellow Fever, Japanese Encephalitis), transmitted predominantly by Aedes and Culex mosquitoes; Togaviridae, which consists of the genus Alphavirus (e.g., Chikungunya, Eastern and Western Equine Encephalitis viruses), also transmitted by Aedes and Culex; Bunyaviridae (recently reorganized), containing viruses like Rift Valley Fever and Oropouche virus, transmitted by mosquitoes and sometimes sandflies; and Reoviridae, which includes the genus Orbivirus (e.g., West Nile and Bluetongue viruses), primarily affecting animals and transmitted by mosquitoes and sandflies. Despite extensive research, effective antiviral treatments for MBVs remain scarce, and current therapies mainly provide symptomatic relief and supportive care. This review examines the viral components and cellular and immune factors involved in the life cycle of MBVs. It also highlights recent advances in antiviral strategies targeting host factors such as lipid metabolism, ion channels, and proteasomes, as well as viral targets like NS2B-NS3 proteases and nonstructural proteins. Additionally, it explores immunomodulatory therapies to enhance antiviral responses and emphasizes the potential of drug repurposing, bioinformatics, artificial intelligence, and deep learning in identifying novel antiviral candidates. Continued research is crucial in mitigating MBVs' impact and preventing future outbreaks.
Collapse
Affiliation(s)
- Ayyaz Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shahid Wahab
- Department of Agriculture, Jeonbuk National University, Jeonju-si, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, 54907, South Korea.
| |
Collapse
|
2
|
Liu J, Quan Y, Tong H, Zhu Y, Shi X, Liu Y, Cheng G. Insights into mosquito-borne arbovirus receptors. CELL INSIGHT 2024; 3:100196. [PMID: 39391003 PMCID: PMC11462183 DOI: 10.1016/j.cellin.2024.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024]
Abstract
The increasing global prevalence of mosquito-borne viruses has emerged as a significant threat to human health and life. Identifying receptors for these viruses is crucial for improving our knowledge of viral pathogenesis and developing effective antiviral strategies. The widespread application of CRISPR-Cas9 screening have led to the discovery of many mosquito-borne virus receptors. The revealed structures of virus-receptor complexes also provide important information for understanding their interaction mechanisms. This review provides a comprehensive summary of both conventional and novel approaches for identifying new viral receptors and the putative entry factors of the most prevalent mosquito-borne viruses within the Flaviviridae, Togaviridae, and Bunyavirales. At the same time, we emphasize the common receptors utilized by these viruses for entry into both vertebrate hosts and mosquito vectors. We discuss promising avenues for developing anti-mosquito-borne viral strategies that target these receptors. Notably, targeting universal receptors of specific mosquito-borne viruses in both vertebrates and mosquitoes offers dual benefits for disease prevention. Additionally, the widespread use of AI-based machine learning and protein structure prediction will accelerate the identification of new viral receptors and provide new avenues for antiviral drug discovery.
Collapse
Affiliation(s)
- Jianying Liu
- 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
| | - Yixin Quan
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
- School of Life Science, Southern University of Science and Technology, Shenzhen, 518052, China
| | - Hua Tong
- 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
| | - 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
| | - Xiaolu Shi
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
| | - Yang Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518000, China
| | - 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
|
3
|
Gao S, Xu H, Li H, Feng X, Zhou J, Guo R, Liang Z, Ding J, Li X, Huang Y, Liu W, Liang S. Identification and functional analysis of C-type lectin from mosquito Aedes albopictus in response to dengue virus infection. Parasit Vectors 2024; 17:375. [PMID: 39232769 PMCID: PMC11373435 DOI: 10.1186/s13071-024-06453-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/16/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND C-type lectins (CTLs) are a large family of proteins with sugar-binding activity. CTLs contain an evolutionarily conserved C-type lectin domain (CTLD) that binds microbial carbohydrates in a calcium-dependent manner, thereby playing a key role in both microbial pathogenesis and innate immune responses. Aedes albopictus is an important vector for transmitting dengue virus (DENV) worldwide. Currently, the molecular characteristics and functions of CTLs in Ae. albopictus are largely unknown. METHODS Transcripts encoding CTL proteins in the Ae. albopictus genome assembly were analyzed via sequence blast. Phylogenetic analysis and molecular characterization were performed to identify the functional domains of the CTLs. Quantitative analysis was performed to determine the gene expression features of CTLs during mosquito development and in different tissues of female adults after blood feeding. In addition, the functional role of CTLs in response to DENV infection was investigated in Ae. albopictus mosquito cells. RESULTS We identified 39 transcripts encoding CTL proteins in the Ae. albopictus transcriptome. Aedes albopictus CTLs are classified into three groups based on the number of CTLDs and the domain architecture. These included 29 CTL-Ss (single-CTLDs), 1 immulectins (dual-CTLD) and 9 CTL-Xs (CTLDs with other domains). Phylogenetic analysis and structural modeling indicated that CTLs in Ae. albopictus are highly conserved with the homologous CTLs in Aedes aegypti. The expression profile assay revealed differential expression patterns of CTLs in both developmental stages and in adult female tissues. Knockdown and overexpression of three CTLs (CTL-S12, S17 and S19) confirmed that they can promote dengue virus infection in Ae. albopictus cells. CONCLUSIONS The CTL genes in Ae. albopictus mosquito and other mosquito species are evolutionarily conserved and exhibit different developmental and tissue expression features. The functional assay indicated that three CTLs in Ae. albopictus mosquitoes are involved in promoting dengue virus infection. Our study revealed that CTLs play important roles in both the physiological processes and viral infection in mosquito vectors.
Collapse
Affiliation(s)
- Sheng Gao
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Haodong Xu
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Hongbo Li
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiao Feng
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jitao Zhou
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Renxian Guo
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zihan Liang
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jinying Ding
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xin Li
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yijia Huang
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Wenquan Liu
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Shaohui Liang
- Department of Medical Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
4
|
Ming Z, Chen Z, Tong H, Zhou X, Feng T, Dai J. Immune functions of C-type lectins in medical arthropods. INSECT SCIENCE 2024; 31:652-662. [PMID: 36661334 DOI: 10.1111/1744-7917.13169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 05/29/2023]
Abstract
C-type lectins (CTLs) are a family of proteins that contain 1 or more carbohydrate-recognition domains (CRDs) and bind to a broad repertoire of ligands in the presence of calcium ions. CTLs play important roles in innate immune defenses against microorganisms by acting as pattern-recognition receptors (PRRs) for invading pathogens, such as bacteria, viruses, and parasites. After binding to pathogen-associated ligands, CTLs mediate immune responses, such as agglutination, phagocytosis, and the activation of phenol oxidase progenitors, thereby clearing pathogens. CTLs are an evolutionarily conserved family found in almost all vertebrates and invertebrates. Medical arthropods can acquire and transmit a range pathogens through various approaches, such as bloodsucking, lancing, and parasitism, thus infecting humans and animals with related diseases, some of which can be life-threatening. Recent studies have shown that lectins are important components of the arthropod immune system and are essential for the immune responses of arthropods to arthropod-borne pathogens. This article reviews the current understanding of the structure, function, and signaling pathways involved in CTLs derived from important medical arthropods.
Collapse
Affiliation(s)
- Zhihao Ming
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, China
| | - Zhiqiang Chen
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, China
| | - Hao Tong
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, China
| | - Xia Zhou
- School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Tingting Feng
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, China
| | - Jianfeng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
5
|
He Y, Miao C, Yang S, Xu C, Liu Y, Zhu X, Wen Y, Wu R, Zhao Q, Huang X, Yan Q, Lang Y, Zhao S, Wang Y, Han X, Cao S, Hu Y, Du S. Sialic acids as attachment factors in mosquitoes mediating Japanese encephalitis virus infection. J Virol 2024; 98:e0195923. [PMID: 38634598 PMCID: PMC11092328 DOI: 10.1128/jvi.01959-23] [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: 12/14/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
The role of Culex mosquitoes in the transmission of Japanese encephalitis virus (JEV) is crucial, yet the mechanisms of JEV infection in these vectors remain unclear. Previous research has indicated that various host factors participate in JEV infection. Herein, we present evidence that mosquito sialic acids enhance JEV infection both in vivo and in vitro. By treating mosquitoes and C6/36 cells with neuraminidase or lectin, the function of sialic acids is effectively blocked, resulting in significant inhibition of JEV infection. Furthermore, knockdown of the sialic acid biosynthesis genes in Culex mosquitoes also leads to a reduction in JEV infection. Moreover, our research revealed that sialic acids play a role in the attachment of JEV to mosquito cells, but not in its internalization. To further explore the mechanisms underlying the promotion of JEV attachment by sialic acids, we conducted immunoprecipitation experiments to confirm the direct binding of sialic acids to the last α-helix in JEV envelope protein domain III. Overall, our study contributes to a molecular comprehension of the interaction between mosquitoes and JEV and offers potential strategies for preventing the dissemination of flavivirus in natural environments.IMPORTANCEIn this study, we aimed to investigate the impact of glycoconjugate sialic acids on mosquito infection with Japanese encephalitis virus (JEV). Our findings demonstrate that sialic acids play a crucial role in enhancing JEV infection by facilitating the attachment of the virus to the cell membrane. Furthermore, our investigation revealed that sialic acids directly bind to the final α-helix in the JEV envelope protein domain III, thereby accelerating virus adsorption. Collectively, our results highlight the significance of mosquito sialic acids in JEV infection within vectors, contributing to a better understanding of the interaction between mosquitoes and JEV.
Collapse
Affiliation(s)
- Yi He
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chang Miao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shiping Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Changhao Xu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuwei Liu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xi Zhu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yifei Lang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Shan Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xinfeng Han
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yajie Hu
- Sichuan Center for Disease Control and Prevention, Chengdu, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Sichuan Science-Observation Experimental Station for Veterinary Drugs and Veterinary Diagnostic Technology, Ministry of Agriculture, Chengdu, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| |
Collapse
|
6
|
Chang YC, Liu WL, Fang PH, Li JC, Liu KL, Huang JL, Chen HW, Kao CF, Chen CH. Effect of C-type lectin 16 on dengue virus infection in Aedes aegypti salivary glands. PNAS NEXUS 2024; 3:pgae188. [PMID: 38813522 PMCID: PMC11134184 DOI: 10.1093/pnasnexus/pgae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/29/2024] [Indexed: 05/31/2024]
Abstract
C-type lectins (CTLs) are a family of carbohydrate-binding proteins and an important component of mosquito saliva. Although CTLs play key roles in immune activation and viral pathogenesis, little is known about their role in regulating dengue virus (DENV) infection and transmission. In this study, we established a homozygous CTL16 knockout Aedes aegypti mutant line using CRISPR/Cas9 to study the interaction between CTL16 and viruses in mosquito vectors. Furthermore, mouse experiments were conducted to confirm the transmission of DENV by CTL16-/- A. aegypti mutants. We found that CTL16 was mainly expressed in the medial lobe of the salivary glands (SGs) in female A. aegypti. CTL16 knockout increased DENV replication and accumulation in the SGs of female A. aegypti, suggesting that CTL16 plays an important role in DENV transmission. We also found a reduced expression of immunodeficiency and Janus kinase/signal transducer and activator of transcription pathway components correlated with increased DENV viral titer, infection rate, and transmission efficiency in the CTL16 mutant strain. The findings of this study provide insights not only for guiding future investigations on the influence of CTLs on immune responses in mosquitoes but also for developing novel mutants that can be used as vector control tools.
Collapse
Affiliation(s)
- Ya-Chen Chang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Pai-Hsiang Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Kun-Lin Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Jau-Ling Huang
- Department of Bioscience Technology, Chang Jung Christian University, Tainan 711301, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Chun-Hong Chen
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 35053, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| |
Collapse
|
7
|
Wu L, Zhang L, Feng S, Chen L, Lin C, Wang G, Zhu Y, Wang P, Cheng G. An evolutionarily conserved ubiquitin ligase drives infection and transmission of flaviviruses. Proc Natl Acad Sci U S A 2024; 121:e2317978121. [PMID: 38593069 PMCID: PMC11032495 DOI: 10.1073/pnas.2317978121] [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: 10/17/2023] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Mosquito-borne flaviviruses such as dengue (DENV) and Zika (ZIKV) cause hundreds of millions of infections annually. The single-stranded RNA genome of flaviviruses is translated into a polyprotein, which is cleaved equally into individual functional proteins. While structural proteins are packaged into progeny virions and released, most of the nonstructural proteins remain intracellular and could become cytotoxic if accumulated over time. However, the mechanism by which nonstructural proteins are maintained at the levels optimal for cellular fitness and viral replication remains unknown. Here, we identified that the ubiquitin E3 ligase HRD1 is essential for flaviviruses infections in both mammalian hosts and mosquitoes. HRD1 directly interacts with flavivirus NS4A and ubiquitylates a conserved lysine residue for ER-associated degradation. This mechanism avoids excessive accumulation of NS4A, which otherwise interrupts the expression of processed flavivirus proteins in the ER. Furthermore, a small-molecule inhibitor of HRD1 named LS-102 effectively interrupts DENV2 infection in both mice and Aedes aegypti mosquitoes, and significantly disturbs DENV transmission from the infected hosts to mosquitoes owing to reduced viremia. Taken together, this study demonstrates that flaviviruses have evolved a sophisticated mechanism to exploit the ubiquitination system to balance the homeostasis of viral proteins for their own advantage and provides a potential therapeutic target to interrupt flavivirus infection and transmission.
Collapse
Affiliation(s)
- Linjuan Wu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen518055, China
| | - Liming Zhang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen518055, China
| | - Lu Chen
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
| | - Cai Lin
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen518000, China
| | - Gang Wang
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
| | - Yibin Zhu
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen518055, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT06030
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing100084, China
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen518000, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen518055, China
- Southwest United Graduate School, Kunming650092, China
| |
Collapse
|
8
|
Yadav K, Saurav GK, Rana VS, Rawat N, Anjali, Jamwal R, Singh OP, Bandyopadhyay A, Rajagopal R. Polyubiquitin protein of Aedes aegypti as an interacting partner of dengue virus envelope protein. MEDICAL AND VETERINARY ENTOMOLOGY 2024; 38:48-58. [PMID: 37807654 DOI: 10.1111/mve.12696] [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: 06/06/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023]
Abstract
Dengue virus (DENV) is an arbovirus that comprises four antigenically different serotypes. Aedes aegypti (Diptera: Culicidae) acts as the principal vector for DENV transmission, and vector control is crucial for dengue fever epidemic management. To design effective vector control strategies, a comprehensive understanding of the insect vector and virus interaction is required. Female Ae. aegypti ingests DENV during the acquisition of a blood meal from an infected human. DENV enters the insect midgut, replicates inside it and reaches the salivary gland for transmitting DENV to healthy humans during the subsequent feeding cycles. DENV must interact with the proteins present in the midgut and salivary glands to gain entry and accomplish successful replication and transmission. Ae. aegypti midgut cDNA library was prepared, and yeast two-hybrid screening was performed against the envelope protein domain III (EDIII) protein of DENV-2. The polyubiquitin protein was selected from the various candidate proteins for subsequent analysis. Polyubiquitin gene was amplified, and the protein was purified in a heterologous expression system for in vitro interaction studies. In vitro pull-down assay presented a clear interaction between polyubiquitin protein and EDIII. To further confirm this interaction, a dot blot assay was employed, and polyubiquitin protein was found to interact with DENV particles. Our results enable us to suggest that polyubiquitin plays an important role in DENV infection within mosquitoes.
Collapse
Affiliation(s)
- Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Nitish Rawat
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Rohit Jamwal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | | | - Anannya Bandyopadhyay
- Protein Homeostasis Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
9
|
Akter R, Tasneem F, Das S, Soma MA, Georgakopoulos-Soares I, Juthi RT, Sazed SA. Approaches of dengue control: vaccine strategies and future aspects. Front Immunol 2024; 15:1362780. [PMID: 38487527 PMCID: PMC10937410 DOI: 10.3389/fimmu.2024.1362780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/08/2024] [Indexed: 03/17/2024] Open
Abstract
Dengue, caused by the dengue virus (DENV), affects millions of people worldwide every year. This virus has two distinct life cycles, one in the human and another in the mosquito, and both cycles are crucial to be controlled. To control the vector of DENV, the mosquito Aedes aegypti, scientists employed many techniques, which were later proved ineffective and harmful in many ways. Consequently, the attention shifted to the development of a vaccine; researchers have targeted the E protein, a surface protein of the virus and the NS1 protein, an extracellular protein. There are several types of vaccines developed so far, such as live attenuated vaccines, recombinant subunit vaccines, inactivated virus vaccines, viral vectored vaccines, DNA vaccines, and mRNA vaccines. Along with these, scientists are exploring new strategies of developing improved version of the vaccine by employing recombinant DNA plasmid against NS1 and also aiming to prevent the infection by blocking the DENV life cycle inside the mosquitoes. Here, we discussed the aspects of research in the field of vaccines until now and identified some prospects for future vaccine developments.
Collapse
Affiliation(s)
- Runa Akter
- Department of Pharmacy, Independent University Bangladesh, Dhaka, Bangladesh
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Faria Tasneem
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Shuvo Das
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | | | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rifat Tasnim Juthi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Saiful Arefeen Sazed
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
10
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
11
|
Mantilla-Granados JS, Castellanos JE, Velandia-Romero ML. A tangled threesome: understanding arbovirus infection in Aedes spp. and the effect of the mosquito microbiota. Front Microbiol 2024; 14:1287519. [PMID: 38235434 PMCID: PMC10792067 DOI: 10.3389/fmicb.2023.1287519] [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: 09/01/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024] Open
Abstract
Arboviral infections transmitted by Aedes spp. mosquitoes are a major threat to human health, particularly in tropical regions but are expanding to temperate regions. The ability of Aedes aegypti and Aedes albopictus to transmit multiple arboviruses involves a complex relationship between mosquitoes and the virus, with recent discoveries shedding light on it. Furthermore, this relationship is not solely between mosquitoes and arboviruses, but also involves the mosquito microbiome. Here, we aimed to construct a comprehensive review of the latest information about the arbovirus infection process in A. aegypti and A. albopictus, the source of mosquito microbiota, and its interaction with the arbovirus infection process, in terms of its implications for vectorial competence. First, we summarized studies showing a new mechanism for arbovirus infection at the cellular level, recently described innate immunological pathways, and the mechanism of adaptive response in mosquitoes. Second, we addressed the general sources of the Aedes mosquito microbiota (bacteria, fungi, and viruses) during their life cycle, and the geographical reports of the most common microbiota in adults mosquitoes. How the microbiota interacts directly or indirectly with arbovirus transmission, thereby modifying vectorial competence. We highlight the complexity of this tripartite relationship, influenced by intrinsic and extrinsic conditions at different geographical scales, with many gaps to fill and promising directions for developing strategies to control arbovirus transmission and to gain a better understanding of vectorial competence. The interactions between mosquitoes, arboviruses and their associated microbiota are yet to be investigated in depth.
Collapse
Affiliation(s)
- Juan S. Mantilla-Granados
- Saneamiento Ecológico, Salud y Medio Ambiente, Universidad El Bosque, Vicerrectoría de Investigaciones, Bogotá, Colombia
| | - Jaime E. Castellanos
- Grupo de Virología, Universidad El Bosque, Vicerrectoría de Investigaciones, Bogotá, Colombia
| | | |
Collapse
|
12
|
Newby ML, Allen JD, Crispin M. Influence of glycosylation on the immunogenicity and antigenicity of viral immunogens. Biotechnol Adv 2024; 70:108283. [PMID: 37972669 PMCID: PMC10867814 DOI: 10.1016/j.biotechadv.2023.108283] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 10/04/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
A key aspect of successful viral vaccine design is the elicitation of neutralizing antibodies targeting viral attachment and fusion glycoproteins that embellish viral particles. This observation has catalyzed the development of numerous viral glycoprotein mimetics as vaccines. Glycans can dominate the surface of viral glycoproteins and as such, the viral glycome can influence the antigenicity and immunogenicity of a candidate vaccine. In one extreme, glycans can form an integral part of epitopes targeted by neutralizing antibodies and are therefore considered to be an important feature of key immunogens within an immunization regimen. In the other extreme, the existence of peptide and bacterially expressed protein vaccines shows that viral glycosylation can be dispensable in some cases. However, native-like glycosylation can indicate native-like protein folding and the presence of conformational epitopes. Furthermore, going beyond native glycan mimicry, in either occupancy of glycosylation sites or the glycan processing state, may offer opportunities for enhancing the immunogenicity and associated protection elicited by an immunogen. Here, we review key determinants of viral glycosylation and how recombinant immunogens can recapitulate these signatures across a range of enveloped viruses, including HIV-1, Ebola virus, SARS-CoV-2, Influenza and Lassa virus. The emerging understanding of immunogen glycosylation and its control will help guide the development of future vaccines in both recombinant protein- and nucleic acid-based vaccine technologies.
Collapse
Affiliation(s)
- Maddy L Newby
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
13
|
Prince BC, Walsh E, Torres TZB, Rückert C. Recognition of Arboviruses by the Mosquito Immune System. Biomolecules 2023; 13:1159. [PMID: 37509194 PMCID: PMC10376960 DOI: 10.3390/biom13071159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to both human and animal health worldwide. These viruses are transmitted through the bites of mosquitoes, ticks, sandflies, or biting midges to humans or animals. In humans, arbovirus infection often results in mild flu-like symptoms, but severe disease and death also occur. There are few vaccines available, so control efforts focus on the mosquito population and virus transmission control. One area of research that may enable the development of new strategies to control arbovirus transmission is the field of vector immunology. Arthropod vectors, such as mosquitoes, have coevolved with arboviruses, resulting in a balance of virus replication and vector immune responses. If this balance were disrupted, virus transmission would likely be reduced, either through reduced replication, or even through enhanced replication, resulting in mosquito mortality. The first step in mounting any immune response is to recognize the presence of an invading pathogen. Recent research advances have been made to tease apart the mechanisms of arbovirus detection by mosquitoes. Here, we summarize what is known about arbovirus recognition by the mosquito immune system, try to generate a comprehensive picture, and highlight where there are still gaps in our current understanding.
Collapse
Affiliation(s)
- Brian C Prince
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Elizabeth Walsh
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Tran Zen B Torres
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
14
|
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: 0.5] [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
|
15
|
Prado Sepulveda CC, Alencar RM, Santana RA, Belém de Souza I, D'Elia GMA, Godoy RSM, Duarte AP, Lopes SCP, de Lacerda MVG, Monteiro WM, Nacif-Pimenta R, Secundino NFC, Koerich LB, Pimenta PFP. Evolution and assembly of Anopheles aquasalis's immune genes: primary malaria vector of coastal Central and South America and the Caribbean Islands. Open Biol 2023; 13:230061. [PMID: 37433331 PMCID: PMC10335856 DOI: 10.1098/rsob.230061] [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: 02/23/2023] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
Anophelines are vectors of malaria, the deadliest disease worldwide transmitted by mosquitoes. The availability of genomic data from various Anopheles species allowed evolutionary comparisons of the immune response genes in search of alternative vector control of the malarial parasites. Now, with the Anopheles aquasalis genome, it was possible to obtain more information about the evolution of the immune response genes. Anopheles aquasalis has 278 immune genes in 24 families or groups. Comparatively, the American anophelines possess fewer genes than Anopheles gambiae s. s., the most dangerous African vector. The most remarkable differences were found in the pathogen recognition and modulation families like FREPs, CLIP and C-type lectins. Even so, genes related to the modulation of the expression of effectors in response to pathogens and gene families that control the production of reactive oxygen species were more conserved. Overall, the results show a variable pattern of evolution in the immune response genes in the anopheline species. Environmental factors, such as exposure to different pathogens and differences in the microbiota composition, could shape the expression of this group of genes. The results presented here will contribute to a better knowledge of the Neotropical vector and open opportunities for malaria control in the endemic-affected areas of the New World.
Collapse
Affiliation(s)
- Cesar Camilo Prado Sepulveda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Rodrigo Maciel Alencar
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Rosa Amélia Santana
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Igor Belém de Souza
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Gigliola Mayra Ayres D'Elia
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Raquel Soares Maia Godoy
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, FIOCRUZ – Belo Horizonte. Minas Gerais, Brazil
| | - Ana Paula Duarte
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Stefanie Costa Pinto Lopes
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Instituto de Pesquisas Leônidas e Maria Deane, Fundação Oswaldo Cruz, Manaus, Amazonas, Brazil
| | - Marcus Vinicius Guimarães de Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Instituto de Pesquisas Leônidas e Maria Deane, Fundação Oswaldo Cruz, Manaus, Amazonas, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | - Wuelton Marcelo Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Rafael Nacif-Pimenta
- Departament of Epidemiology of Microbial Disease, Yale School of Public Health, New Haven, CT, USA
| | - Nágila Francinete Costa Secundino
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, FIOCRUZ – Belo Horizonte. Minas Gerais, Brazil
| | - Leonardo Barbosa Koerich
- Departamento de Parasitologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Filemon Paolucci Pimenta
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Fundação de Medicina Tropical Heitor Vieira Dourado, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, FIOCRUZ – Belo Horizonte. Minas Gerais, Brazil
| |
Collapse
|
16
|
Bi J, Wang Y, Gao R, Liu P, Jiang Y, Gao L, Li B, Song Q, Ning M. Functional Analysis of a CTL-X-Type Lectin CTL16 in Development and Innate Immunity of Tribolium castaneum. Int J Mol Sci 2023; 24:10700. [PMID: 37445878 DOI: 10.3390/ijms241310700] [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: 05/10/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
C-type lectins (CTLs) are a class of proteins containing carbohydrate recognition domains (CRDs), which are characteristic modules that recognize various glycoconjugates and function primarily in immunity. CTLs have been reported to affect growth and development and positively regulate innate immunity in Tribolium castaneum. However, the regulatory mechanisms of TcCTL16 proteins are still unclear. Here, spatiotemporal analyses displayed that TcCTL16 was highly expressed in late pupae and early adults. TcCTL16 RNA interference in early larvae shortened their body length and narrowed their body width, leading to the death of 98% of the larvae in the pupal stage. Further analysis found that the expression level of muscle-regulation-related genes, including cut, vestigial, erect wing, apterous, and spalt major, and muscle-composition-related genes, including Myosin heavy chain and Myosin light chain, were obviously down-regulated after TcCTL16 silencing in T. castaneum. In addition, the transcription of TcCTL16 was mainly distributed in the hemolymph. TcCTL16 was significantly upregulated after challenges with lipopolysaccharides, peptidoglycans, Escherichia coli, and Staphylococcus aureus. Recombinant CRDs of TcCTL16 bind directly to the tested bacteria (except Bacillus subtilis); they also induce extensive bacterial agglutination in the presence of Ca2+. On the contrary, after TcCTL16 silencing in the late larval stage, T. castaneum were able to develop normally. Moreover, the transcript levels of seven antimicrobial peptide genes (attacin2, defensins1, defensins2, coleoptericin1, coleoptericin2, cecropins2, and cecropins3) and one transcription factor gene (relish) were significantly increased under E. coli challenge and led to an increased survival rate of T. castaneum when infected with S. aureus or E. coli, suggesting that TcCTL16 deficiency could be compensated for by increasing AMP expression via the IMD pathways in T. castaneum. In conclusion, this study found that TcCTL16 could be involved in developmental regulation in early larvae and compensate for the loss of CTL function by regulating the expression of AMPs in late larvae, thus laying a solid foundation for further studies on T. castaneum CTLs.
Collapse
Affiliation(s)
- Jingxiu Bi
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Yutao Wang
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Rui Gao
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Pingxiang Liu
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Yuying Jiang
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Lei Gao
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA
| | - Mingxiao Ning
- Laboratory of Quality and Safety Risk Assessment for Agro-Products of the Ministry of Agriculture (Jinan), Institute of Quality Standard and Testing Technology for Agro-Products, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| |
Collapse
|
17
|
Yadav K, Rana VS, Anjali, Saurav GK, Rawat N, Kumar A, Sunil S, Singh OP, Rajagopal R. Mucin Protein of Aedes aegypti Interacts with Dengue Virus 2 and Influences Viral Infection. Microbiol Spectr 2023; 11:e0250322. [PMID: 36847498 PMCID: PMC10101019 DOI: 10.1128/spectrum.02503-22] [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: 07/01/2022] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
Dengue, caused by dengue virus (DENV), is the most prevalent vector-borne viral disease, posing a serious health concern to 2.5 billion people worldwide. DENV is primarily transmitted among humans by its mosquito vector Aedes aegypti; hence, the identification of a novel dengue virus receptor in mosquitoes is critical for the development of new anti-mosquito measures. In the current study, we have identified peptides which potentially interact with the surface of the virion particles and facilitate virus infection and movement during their life cycle in the mosquito vector. To identify these candidate proteins, we performed phage-display library screening against domain III of the envelope protein (EDIII), which plays an essential role during host cell receptor binding for viral entry. The mucin protein, which shared sequence similarity with the peptide identified in the screening, was cloned, expressed, and purified for in vitro interaction studies. Using in vitro pulldown and virus overlay protein-binding assay (VOPBA), we confirmed the positive interaction of mucin with purified EDIII and whole virion particles. Finally, blocking of mucin protein with anti-mucin antibodies partially reduced DENV titers in infected mosquitos. Moreover, mucin protein was found to be localized in the midgut of Ae. aegypti. IMPORTANCE Identification of interacting protein partners of DENV in the insect vector Aedes aegypti is crucial for designing vector control-based strategies and for understanding the molecular mechanism DENV uses to modulate the host, gain entry, and survive successfully. Similar proteins can be used in generating transmission-blocking vaccines.
Collapse
Affiliation(s)
- Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Vipin Singh Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Anjali
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Gunjan Kumar Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
- Department of Zoology, Rajiv Gandhi University, Doimukh, Arunachal Pradesh, India
| | - Nitish Rawat
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| | - Ankit Kumar
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Disease Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Om P. Singh
- National Institute of Malaria Research, New Delhi, India
| | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India
| |
Collapse
|
18
|
Stanley D, Haas E, Kim Y. Beyond Cellular Immunity: On the Biological Significance of Insect Hemocytes. Cells 2023; 12:cells12040599. [PMID: 36831266 PMCID: PMC9954174 DOI: 10.3390/cells12040599] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Insect immunity is assorted into humoral and cellular immune reactions. Humoral reactions involve the regulated production of anti-microbial peptides, which directly kill microbial invaders at the membrane and intracellular levels. In cellular immune reactions, millions of hemocytes are mobilized to sites of infection and replaced by hematopoiesis at a high biological cost after the immune defense. Here, we considered that the high biological costs of maintaining and replacing hemocytes would be a better investment if hemocytes carried out meaningful biological actions unrelated to cellular immunity. This idea allows us to treat a set of 10 hemocyte actions that are not directly involved in immunity, some of which, so far, are known only in Drosophila melanogaster. These include (1) their actions in molting and development, (2) in surviving severe hypoxia, (3) producing phenoloxidase precursor and its actions beyond immunity, (4) producing vitellogenin in a leafhopper, (5) recognition and responses to cancer in Drosophila, (6) non-immune actions in Drosophila, (7) clearing apoptotic cells during development of the central nervous system, (8) developing hematopoietic niches in Drosophila, (9) synthesis and transport of a lipoprotein, and (10) hemocyte roles in iron transport. We propose that the biological significance of hemocytes extends considerably beyond immunity.
Collapse
Affiliation(s)
- David Stanley
- Biological Control of Insect Research Laboratory, USDA/ARS, 1503 S Providence Road, Columbia, MO 65203, USA
- Correspondence: (D.S.); (Y.K.)
| | - Eric Haas
- Department of Chemistry and Biochemistry, Creighton University, 2500 California Plaza, Omaha, NE 68178, USA
| | - Yonggyun Kim
- Department of Plant Medicals, Andong National University, Andong 36729, Republic of Korea
- Correspondence: (D.S.); (Y.K.)
| |
Collapse
|
19
|
Chen L, Zhang X, Guo X, Peng W, Zhu Y, Wang Z, Yu X, Shi H, Li Y, Zhang L, Wang L, Wang P, Cheng G. Neighboring mutation-mediated enhancement of dengue virus infectivity and spread. EMBO Rep 2022; 23:e55671. [PMID: 36197120 PMCID: PMC9638853 DOI: 10.15252/embr.202255671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 10/07/2023] Open
Abstract
Frequent turnover of dengue virus (DENV) clades is one of the major forces driving DENV persistence and prevalence. In this study, we assess the fitness advantage of nine stable substitutions within the envelope (E) protein of DENV serotypes. Two tandem neighboring substitutions, threonine to lysine at the 226th (T226K) and glycine to glutamic acid at the 228th (G228E) residues in the DENV2 Asian I genotype, enhance virus infectivity in either mosquitoes or mammalian hosts, thereby promoting clades turnover and dengue epidemics. Mechanistic studies indicate that the substitution-mediated polarity changes in these two residues increase the binding affinity of E for host C-type lectins. Accordingly, we predict that a G228E substitution could potentially result in a forthcoming epidemic of the DENV2 Cosmopolitan genotype. Investigations into the substitutions associated with DENV fitness in hosts may offer mechanistic insights into dengue prevalence, thus providing a warning of potential epidemics in the future.
Collapse
Affiliation(s)
- Lu Chen
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Xianwen Zhang
- Institute of Infectious DiseasesShenzhen Bay LaboratoryShenzhenChina
| | - Xuan Guo
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Wenyu Peng
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Yibin Zhu
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Zhaoyang Wang
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Xi Yu
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Huicheng Shi
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Yuhan Li
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Liming Zhang
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
| | - Lei Wang
- Institute of Infectious DiseasesShenzhen Bay LaboratoryShenzhenChina
| | - Penghua Wang
- Department of Immunology, School of Medicinethe University of Connecticut Health CenterFarmingtonCTUSA
| | - Gong Cheng
- Tsinghua‐Peking Joint Center for Life Sciences, School of MedicineTsinghua UniversityBeijingChina
- Institute of Infectious DiseasesShenzhen Bay LaboratoryShenzhenChina
| |
Collapse
|
20
|
Peinado RDS, Eberle RJ, Arni RK, Coronado MA. A Review of Omics Studies on Arboviruses: Alphavirus, Orthobunyavirus and Phlebovirus. Viruses 2022; 14:2194. [PMID: 36298749 PMCID: PMC9607206 DOI: 10.3390/v14102194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Since the intricate and complex steps in pathogenesis and host-viral interactions of arthropod-borne viruses or arboviruses are not completely understood, the multi-omics approaches, which encompass proteomics, transcriptomics, genomics and metabolomics network analysis, are of great importance. We have reviewed the omics studies on mosquito-borne viruses of the Togaviridae, Peribuyaviridae and Phenuiviridae families, specifically for Chikungunya, Mayaro, Oropouche and Rift Valley Fever viruses. Omics studies can potentially provide a new perspective on the pathophysiology of arboviruses, contributing to a better comprehension of these diseases and their effects and, hence, provide novel insights for the development of new antiviral drugs or therapies.
Collapse
Affiliation(s)
- Rafaela dos S. Peinado
- Multiuser Center for Biomolecular Innovation, Department of Physics, Sao Paulo State University, Sao Jose do Rio Preto 15054-000, SP, Brazil
| | - Raphael J. Eberle
- Institute of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, Sao Paulo State University, Sao Jose do Rio Preto 15054-000, SP, Brazil
| | - Mônika A. Coronado
- Institute of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
21
|
Cardoso-Jaime V, Tikhe CV, Dong S, Dimopoulos G. The Role of Mosquito Hemocytes in Viral Infections. Viruses 2022; 14:v14102088. [PMID: 36298644 PMCID: PMC9608948 DOI: 10.3390/v14102088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Insect hemocytes are the only immune cells that can mount a humoral and cellular immune response. Despite the critical involvement of hemocytes in immune responses against bacteria, fungi, and parasites in mosquitoes, our understanding of their antiviral potential is still limited. It has been shown that hemocytes express humoral factors such as TEP1, PPO, and certain antimicrobial peptides that are known to restrict viral infections. Insect hemocytes also harbor the major immune pathways, such as JAK/STAT, TOLL, IMD, and RNAi, which are critical for the control of viral infection. Recent research has indicated a role for hemocytes in the regulation of viral infection through RNA interference and autophagy; however, the specific mechanism by which this regulation occurs remains uncharacterized. Conversely, some studies have suggested that hemocytes act as agonists of arboviral infection because they lack basal lamina and circulate throughout the whole mosquito, likely facilitating viral dissemination to other tissues such as salivary glands. In addition, hemocytes produce arbovirus agonist factors such as lectins, which enhance viral infection. Here, we summarize our current understanding of hemocytes’ involvement in viral infections.
Collapse
|
22
|
Abduljalil JM, Abd Al Galil FM. Molecular pathogenesis of dengue virus infection in Aedes mosquitoes. JOURNAL OF INSECT PHYSIOLOGY 2022; 138:104367. [PMID: 35131236 DOI: 10.1016/j.jinsphys.2022.104367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 06/14/2023]
Abstract
Aedes mosquitoes are implicated in the transmission of several viruses, including Dengue virus (DENV) to millions of people worldwide. The global expansion of Aedes mosquitos'habitats creates a desperate need for control mechanisms with minimum negative effects. Deciphering the molecular interactions between DENV and its vector is a promising field to develop such efficient control strategies. As soon as the viremic blood is ingested by the mosquito, DENV is encountered by different innate immunity responses. During the past three decades, different pathways of innate immunity have been identified in Aedes spp. Recognition of viral molecular patterns, including viral RNA, and vector attempts to resist DENV infection are the most important defense mechanisms. Crosstalk between innate immune pathways and redundancy of anti-DENV responses become more evident as research progresses. The viral evasion and repression of vector immune response are increasingly being discovered. Such viral strategies are potential targets to be disrupted in order to limit DENV infection and spread. Vector-related non-immune factors such as gut microbiota can also be tapped for efficient control of DENV infection in Aedes mosquito's populations without affecting their fitness. Current trends in controlling DENV in its vector are exploring the potentials of using genetically engineered mosquitoes via RNA-based systems to degrade DENV genome once released into the midgut cells cytoplasm at the early phase of the infection.
Collapse
Affiliation(s)
- Jameel M Abduljalil
- Department of Biological Sciences, Faculty of Applied Sciences, Thamar University, B.O. Box: 87246, Yemen.
| | - Fahd M Abd Al Galil
- Department of Biological Sciences, Faculty of Applied Sciences, Thamar University, B.O. Box: 87246, Yemen; Department of Biology, Faculty of Sciences, University of Bisha, B.O. Box: 551, Bisha, Saudi Arabia.
| |
Collapse
|
23
|
Schön K, Lindenwald DL, Monteiro JT, Glanz J, Jung K, Becker SC, Lepenies B. Vector and Host C-Type Lectin Receptor (CLR)-Fc Fusion Proteins as a Cross-Species Comparative Approach to Screen for CLR-Rift Valley Fever Virus Interactions. Int J Mol Sci 2022; 23:ijms23063243. [PMID: 35328665 PMCID: PMC8954825 DOI: 10.3390/ijms23063243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus endemic to Africa and the Arabian Peninsula, which causes diseases in humans and livestock. C-type lectin receptors (CLRs) represent a superfamily of pattern recognition receptors that were reported to interact with diverse viruses and contribute to antiviral immune responses but may also act as attachment factors or entry receptors in diverse species. Human DC-SIGN and L-SIGN are known to interact with RVFV and to facilitate viral host cell entry, but the roles of further host and vector CLRs are still unknown. In this study, we present a CLR–Fc fusion protein library to screen RVFV–CLR interaction in a cross-species approach and identified novel murine, ovine, and Aedes aegypti RVFV candidate receptors. Furthermore, cross-species CLR binding studies enabled observations of the differences and similarities in binding preferences of RVFV between mammalian CLR homologues, as well as more distant vector/host CLRs.
Collapse
Affiliation(s)
- Kathleen Schön
- Institute for Parasitology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany;
- Institute for Immunology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (D.L.L.); (J.T.M.)
| | - Dimitri L. Lindenwald
- Institute for Immunology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (D.L.L.); (J.T.M.)
| | - João T. Monteiro
- Institute for Immunology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (D.L.L.); (J.T.M.)
| | - Julien Glanz
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (J.G.); (K.J.)
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (J.G.); (K.J.)
| | - Stefanie C. Becker
- Institute for Parasitology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany;
- Correspondence: (S.C.B.); (B.L.)
| | - Bernd Lepenies
- Institute for Immunology & Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hanover, Germany; (D.L.L.); (J.T.M.)
- Correspondence: (S.C.B.); (B.L.)
| |
Collapse
|
24
|
Chowdhury A, Modahl CM, Missé D, Kini RM, Pompon J. High resolution proteomics of Aedes aegypti salivary glands infected with either dengue, Zika or chikungunya viruses identify new virus specific and broad antiviral factors. Sci Rep 2021; 11:23696. [PMID: 34880409 PMCID: PMC8654903 DOI: 10.1038/s41598-021-03211-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022] Open
Abstract
Arboviruses such as dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses infect close to half a billion people per year, and are primarily transmitted through Aedes aegypti bites. Infection-induced changes in mosquito salivary glands (SG) influence transmission by inducing antiviral immunity, which restricts virus replication in the vector, and by altering saliva composition, which influences skin infection. Here, we profiled SG proteome responses to DENV serotype 2 (DENV2), ZIKV and CHIKV infections by using high-resolution isobaric-tagged quantitative proteomics. We identified 218 proteins with putative functions in immunity, blood-feeding or related to the cellular machinery. We observed that 58, 27 and 29 proteins were regulated by DENV2, ZIKV and CHIKV infections, respectively. While the regulation patterns were mostly virus-specific, we separately depleted four uncharacterized proteins that were upregulated by all three viral infections to determine their effects on these viral infections. Our study suggests that gamma-interferon responsive lysosomal thiol-like (GILT-like) has an anti-ZIKV effect, adenosine deaminase (ADA) has an anti-CHIKV effect, salivary gland surface protein 1 (SGS1) has a pro-ZIKV effect and salivary gland broad-spectrum antiviral protein (SGBAP) has an antiviral effect against all three viruses. The comprehensive description of SG responses to three global pathogenic viruses and the identification of new restriction factors improves our understanding of the molecular mechanisms influencing transmission.
Collapse
Affiliation(s)
- Avisha Chowdhury
- grid.4280.e0000 0001 2180 6431Department of Biological Science, National University of Singapore, Singapore, Singapore ,grid.428397.30000 0004 0385 0924Present Address: Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Cassandra M. Modahl
- grid.4280.e0000 0001 2180 6431Department of Biological Science, National University of Singapore, Singapore, Singapore ,grid.48004.380000 0004 1936 9764Present Address: Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
| | - Dorothée Missé
- grid.462603.50000 0004 0382 3424MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - R. Manjunatha Kini
- grid.4280.e0000 0001 2180 6431Department of Biological Science, National University of Singapore, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Julien Pompon
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France. .,Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore. .,MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France.
| |
Collapse
|
25
|
Yu X, Shan C, Zhu Y, Ma E, Wang J, Wang P, Shi PY, Cheng G. A mutation-mediated evolutionary adaptation of Zika virus in mosquito and mammalian host. Proc Natl Acad Sci U S A 2021; 118:e2113015118. [PMID: 34620704 PMCID: PMC8545446 DOI: 10.1073/pnas.2113015118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 01/18/2023] Open
Abstract
Zika virus (ZIKV) caused millions of infections during its rapid and expansive spread from Asia to the Americas from 2015 to 2017. Here, we compared the infectivity of ZIKV mutants with individual stable substitutions which emerged throughout the Asian ZIKV lineage and were responsible for the explosive outbreaks in the Americas. A threonine (T) to alanine (A) mutation at the 106th residue of the ZIKV capsid (C) protein facilitated the transmission by its mosquito vector, as well as infection in both human cells and immunodeficient mice. A mechanistic study showed that the T106A substitution rendered the C a preferred substrate for the NS2B-NS3 protease, thereby facilitating the maturation of structural proteins and the formation of infectious viral particles. Over a complete "mosquito-mouse-mosquito" cycle, the ZIKV C-T106A mutant showed a higher prevalence of mosquito infection than did the preepidemic strain, thus promoting ZIKV dissemination. Our results support the contribution of this evolutionary adaptation to the occasional widespread reemergence of ZIKV in nature.
Collapse
Affiliation(s)
- Xi Yu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen 518000, China
- Shenzhen Center for Disease Control and Prevention, Institute of Pathogenic Organisms, Shenzhen 518055, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555
| | - Yibin Zhu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen 518000, China
- Shenzhen Center for Disease Control and Prevention, Institute of Pathogenic Organisms, Shenzhen 518055, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jinglin Wang
- Yunnan Tropical and Subtropical Animal Viral Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming 650224, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, CT 06030
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China;
- Shenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen 518000, China
- Shenzhen Center for Disease Control and Prevention, Institute of Pathogenic Organisms, Shenzhen 518055, China
| |
Collapse
|
26
|
Wang WH, Urbina AN, Lin CY, Yang ZS, Assavalapsakul W, Thitithanyanont A, Lu PL, Chen YH, Wang SF. Targets and strategies for vaccine development against dengue viruses. Biomed Pharmacother 2021; 144:112304. [PMID: 34634560 DOI: 10.1016/j.biopha.2021.112304] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022] Open
Abstract
Dengue virus (DENV) is a global health threat causing about half of the worldwide population to be at risk of infection, especially the people living in tropical and subtropical area. Although the dengue disease caused by dengue virus (DENV) is asymptomatic and self-limiting in most people with first infection, increased severe dengue symptoms may be observed in people with heterotypic secondary DENV infection. Since there is a lack of specific antiviral medication, the development of dengue vaccines is critical in the prevention and control this disease. Several targets and strategies in the development of dengue vaccine have been demonstrated. Currently, Dengvaxia, a live-attenuated chimeric yellow-fever/tetravalent dengue vaccine (CYD-TDV) developed by Sanofi Pasteur, has been licensed and approved for clinical use in some countries. However, this vaccine has demonstrated low efficacy in children and dengue-naïve individuals and also increases the risk of severe dengue in young vaccinated recipients. Accordingly, many novel strategies for the dengue vaccine are under investigation and development. Here, we conducted a systemic literature review according to PRISMA guidelines to give a concise overview of various aspects of the vaccine development process against DENVs, mainly targeting five potential strategies including live attenuated vaccine, inactivated virus vaccine, recombinant subunit vaccine, viral-vector vaccine, and DNA vaccine. This study offers the comprehensive view of updated information and current progression of immunogen selection as well as strategies of vaccine development against DENVs.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Aspiro Nayim Urbina
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Yen Lin
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Zih-Syuan Yang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Po-Liang Lu
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yen-Hsu Chen
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
27
|
Liu D, Xiao X, Zhou P, Zheng H, Li Y, Jin H, Jongkaewwattana A, Luo R. Glycosylation on envelope glycoprotein of duck Tembusu virus affects virus replication in vitro and contributes to the neurovirulence and pathogenicity in vivo. Virulence 2021; 12:2400-2414. [PMID: 34506259 PMCID: PMC8437475 DOI: 10.1080/21505594.2021.1974329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Duck Tembusu virus (DTMUV), an emergent flavivirus, causes domestic waterfowls to suffer from severe egg-drop syndrome and fatal encephalitis, greatly threatens duck production globally. Like other mosquito-borne flaviviruses, the envelope (E) protein of all DTMUV strains was N-glycosylated at the amino acid position 154. Thus far, the biological roles of DTMUV E glycosylation have remained largely unexplored. Herein, we demonstrated the key roles of E glycosylation in the replication and pathogenicity of DTMUV in ducks by characterizing the reverse-genetics-derived DTMUV wild-type MC strain and MC bearing mutations (N154Q and N154I) that abolish the E glycosylation. Our data showed that the disruption of E glycosylation could substantially impair virus attachment, entry, and infectivity in DEFs and C6/36 cells. Notably, ducks inoculated intracerebrally with the wild-type virus exhibited severe disease onset. In contrast, those inoculated with mutant viruses were mildly affected as manifested by minimal weight loss, no mortality, lower viral loads in the various tissues, and reduced brain lesions. Attenuated phenotypes of the mutant viruses might be partly associated with lower inflammatory cytokines expression in the brains of infected ducks. Our study offers the first evidence that E glycosylation is vital for DTMUV replication, pathogenicity, and neurovirulence in vivo.
Collapse
Affiliation(s)
- Dejian Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Xuyao Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Huijun Zheng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Yaqian Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (Biotec), National Science and Technology Development Agency (Nstda), Klong Nueng, Pathum Thani Thailand
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| |
Collapse
|
28
|
Marin-Lopez A, Jiang J, Wang Y, Cao Y, MacNeil T, Hastings AK, Fikrig E. Aedes aegypti SNAP and a calcium transporter ATPase influence dengue virus dissemination. PLoS Negl Trop Dis 2021; 15:e0009442. [PMID: 34115766 PMCID: PMC8195420 DOI: 10.1371/journal.pntd.0009442] [Citation(s) in RCA: 4] [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: 12/08/2020] [Accepted: 05/04/2021] [Indexed: 11/19/2022] Open
Abstract
Dengue virus (DENV) is a flavivirus that causes marked human morbidity and mortality worldwide, and is transmitted to humans by Aedes aegypti mosquitoes. Habitat expansion of Aedes, mainly due to climate change and increasing overlap between urban and wild habitats, places nearly half of the world's population at risk for DENV infection. After a bloodmeal from a DENV-infected host, the virus enters the mosquito midgut. Next, the virus migrates to, and replicates in, other tissues, like salivary glands. Successful viral transmission occurs when the infected mosquito takes another blood meal on a susceptible host and DENV is released from the salivary gland via saliva into the skin. During viral dissemination in the mosquito and transmission to a new mammalian host, DENV interacts with a variety of vector proteins, which are uniquely important during each phase of the viral cycle. Our study focuses on the interaction between DENV particles and protein components in the A. aegypti vector. We performed a mass spectrometry assay where we identified a set of A. aegypti salivary gland proteins which potentially interact with the DENV virion. Using dsRNA to silence gene expression, we analyzed the role of these proteins in viral infectivity. Two of these candidates, a synaptosomal-associated protein (AeSNAP) and a calcium transporter ATPase (ATPase) appear to play a role in viral replication both in vitro and in vivo, observing a ubiquitous expression of these proteins in the mosquito. These findings suggest that AeSNAP plays a protective role during DENV infection of mosquitoes and that ATPase protein is required for DENV during amplification within the vector.
Collapse
Affiliation(s)
- Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Junjun Jiang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuchen Wang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- State Key Laboratory of Virology, College of Life Science, Wuhan University, Wuhan, Hubei, China
| | - Yongguo Cao
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Clinical Veterinary Medicine, and Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tyler MacNeil
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Andrew K. Hastings
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
29
|
Weng SC, Li HH, Li JC, Liu WL, Chen CH, Shiao SH. A Thioester-Containing Protein Controls Dengue Virus Infection in Aedes aegypti Through Modulating Immune Response. Front Immunol 2021; 12:670122. [PMID: 34054842 PMCID: PMC8155531 DOI: 10.3389/fimmu.2021.670122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023] Open
Abstract
Complement-like proteins in arthropods defend against invading pathogens in the early phases of infection. Thioester-containing proteins (TEPs), which exhibit high similarity to mammalian complement C3, are thought to play a key role in the innate immunity of arthropods. We identified and characterized anti-dengue virus (DENV) host factors, in particular complement-like proteins, in the mosquito Aedes aegypti. Our results indicate that TEP1 limits DENV infection in Ae. aegypti. We showed that TEP1 transcription is highly induced in mosquitoes following DENV infection. Silencing TEP1 resulted in the up-regulation of viral RNA and proteins. In addition, the production of infectious virus particles increased in the absence of TEP1. We generated a transgenic mosquito line with a TEP1 loss-of-function phenotype under a blood meal-inducible promoter. We showed that viral protein and titers increased in transgenic mosquitoes after an infectious blood meal. Interestingly, expression of transcription factor Rel2 and certain anti-microbial peptides (AMPs) were inhibited in transgenic mosquitoes. Overall, our results suggest that TEP1 regulates the immune response and consequently controls the replication of dengue virus in mosquitoes. This finding provides new insight into the molecular mechanisms of mosquito host factors in the regulation of DENV replication.
Collapse
Affiliation(s)
- Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsing-Han Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Wei-Liang Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Sánchez-González G, Belak ZR, Lozano L, Condé R. Probability of consolidation constrains novel serotype emergence in dengue fever virus. PLoS One 2021; 16:e0248765. [PMID: 33819302 PMCID: PMC8021166 DOI: 10.1371/journal.pone.0248765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
Since their first sequencing 40 years ago, Dengue virus (DENV) genotypes have shown extreme coherence regarding the serotype class they encode. Considering that DENV is a ribonucleic acid (RNA) virus with a high mutation rate, this behavior is intriguing. Here, we explore the effect of various parameters on likelihood of new serotype emergence. In order to determine the time scales of such an event, we used a Timed Markov Transmission Model to explore the influences of sylvatic versus peri-urban transmission, viral mutation rate, and vertical transmission on the probabilities of novel serotype emergence. We found that around 1 000 years are required for a new serotype to emerge, consistent with phylogenetic analysis of extant dengue serotypes. Furthermore, we show that likelihood of establishing chains of mosquito-human-mosquito infection, known as consolidation, is the primary factor which constrains novel serotype emergence. Our work illustrates the restrictions on and provides a mechanistic explanation for the low probability of novel dengue virus serotype emergence and the low number of observed DENV serotypes.
Collapse
Affiliation(s)
- Gilberto Sánchez-González
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | | | - Luis Lozano
- Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Renaud Condé
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| |
Collapse
|
31
|
Cheng L, Liu WL, Tsou YT, Li JC, Chien CH, Su MP, Liu KL, Huang YL, Wu SC, Tsai JJ, Hsieh SL, Chen CH. Transgenic Expression of Human C-Type Lectin Protein CLEC18A Reduces Dengue Virus Type 2 Infectivity in Aedes aegypti. Front Immunol 2021; 12:640367. [PMID: 33767710 PMCID: PMC7985527 DOI: 10.3389/fimmu.2021.640367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/17/2021] [Indexed: 01/15/2023] Open
Abstract
The C-type lectins, one family of lectins featuring carbohydrate binding domains which participate in a variety of bioprocesses in both humans and mosquitoes, including immune response, are known to target DENV. A human C-type lectin protein CLEC18A in particular shows extensive glycan binding abilities and correlates with type-I interferon expression, making CLEC18A a potential player in innate immune responses to DENV infection; this potential may provide additional regulatory point in improving mosquito immunity. Here, we established for the first time a transgenic Aedes aegypti line that expresses human CLEC18A. This expression enhanced the Toll immune pathway responses to DENV infection. Furthermore, viral genome and virus titers were reduced by 70% in the midgut of transgenic mosquitoes. We found significant changes in the composition of the midgut microbiome in CLEC18A expressing mosquitoes, which may result from the Toll pathway enhancement and contribute to DENV inhibition. Transgenic mosquito lines offer a compelling option for studying DENV pathogenesis, and our analyses indicate that modifying the mosquito immune system via expression of a human immune gene can significantly reduce DENV infection.
Collapse
Affiliation(s)
- Lie Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Yun-Ting Tsou
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Hao Chien
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Matthew P Su
- Department of Biological Science, Nagoya University, Nagoya, Japan
| | - Kun-Lin Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Ya-Lang Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Cheng Wu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Jih-Jin Tsai
- Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
32
|
Bakhshi H, Fazlalipour M, Dadgar-Pakdel J, Zakeri S, Raz A, Failloux AB, Dinparast Djadid N. Developing a Vaccine to Block West Nile Virus Transmission: In Silico Studies, Molecular Characterization, Expression, and Blocking Activity of Culex pipiens mosGCTL-1. Pathogens 2021; 10:pathogens10020218. [PMID: 33671430 PMCID: PMC7921969 DOI: 10.3390/pathogens10020218] [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: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Mosquito galactose-specific C-type lectins (mosGCTLs), such as mosGCTL-1, act as ligands to facilitate the invasion of flaviviruses like West Nile virus (WNV). WNV interacts with the mosGCTL-1 of Aedes aegypti (Culicidae) and facilitates the invasion of this virus. Nevertheless, there is no data about the role of mosGCTL-1 as a transmission-blocking vaccine candidate in Culex pipiens, the most abundant Culicinae mosquito in temperate regions. METHODS Adult female Cx. pipiens mosquitoes were experimentally infected with a WNV infectious blood meal, and the effect of rabbit anti-rmosGCTL-1 antibodies on virus replication was evaluated. Additionally, in silico studies such as the prediction of protein structure, homology modeling, and molecular interactions were carried out. RESULTS We showed a 30% blocking activity of Cx. pipiens mosGCTL-1 polyclonal antibodies (compared to the 10% in the control group) with a decrease in infection rates in mosquitoes at day 5 post-infection, suggesting that there may be other proteins in the midgut of Cx. pipiens that could act as cooperative-receptors for WNV. In addition, docking results revealed that WNV binds with high affinity, to the Culex mosquito lectin receptors. CONCLUSIONS Our results do not support the idea that mosGCTL-1 of Cx. pipiens primarily interacts with WNV to promote viral infection, suggesting that other mosGCTLs may act as primary infection factors in Cx. pipiens.
Collapse
Affiliation(s)
- Hasan Bakhshi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran; (H.B.); (J.D.-P.); (S.Z.)
| | - Mehdi Fazlalipour
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Ref Lab), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran;
| | - Javad Dadgar-Pakdel
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran; (H.B.); (J.D.-P.); (S.Z.)
- Trauma Research Center, Sina Hospital, Tehran University of Medical Sciences, Hassan Abad Square, Imam Khomeini Avenue, Tehran 1136746911, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran; (H.B.); (J.D.-P.); (S.Z.)
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran; (H.B.); (J.D.-P.); (S.Z.)
- Correspondence: (A.R.); (A.-B.F.); (N.D.D.); Tel.: +98-(0)21-64-11-24-62 (A.R.); +33-(0)1-40-61-36-17 (A.-B.F.); +98-(0)21-64-11-24-62 (N.D.D.)
| | - Anna-Bella Failloux
- Institut Pasteur, Department of Virology, Arboviruses and Insect Vectors, 25 rue Dr. Roux, CEDEX 15, 75724 Paris, France
- Correspondence: (A.R.); (A.-B.F.); (N.D.D.); Tel.: +98-(0)21-64-11-24-62 (A.R.); +33-(0)1-40-61-36-17 (A.-B.F.); +98-(0)21-64-11-24-62 (N.D.D.)
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran; (H.B.); (J.D.-P.); (S.Z.)
- Correspondence: (A.R.); (A.-B.F.); (N.D.D.); Tel.: +98-(0)21-64-11-24-62 (A.R.); +33-(0)1-40-61-36-17 (A.-B.F.); +98-(0)21-64-11-24-62 (N.D.D.)
| |
Collapse
|
33
|
Núñez AI, Esteve-Codina A, Gómez-Garrido J, Brustolin M, Talavera S, Berdugo M, Dabad M, Alioto T, Bensaid A, Busquets N. Alteration in the Culex pipiens transcriptome reveals diverse mechanisms of the mosquito immune system implicated upon Rift Valley fever phlebovirus exposure. PLoS Negl Trop Dis 2020; 14:e0008870. [PMID: 33301456 PMCID: PMC7755283 DOI: 10.1371/journal.pntd.0008870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/22/2020] [Accepted: 10/10/2020] [Indexed: 01/02/2023] Open
Abstract
Rift Valley fever phlebovirus (RVFV) causes an emerging zoonotic disease and is mainly transmitted by Culex and Aedes mosquitoes. While Aedes aegypti-dengue virus (DENV) is the most studied model, less is known about the genes involved in infection-responses in other mosquito-arboviruses pairing. The main objective was to investigate the molecular responses of Cx. pipiens to RVFV exposure focusing mainly on genes implicated in innate immune responses. Mosquitoes were fed with blood spiked with RVFV. The fully-engorged females were pooled at 3 different time points: 2 hours post-exposure (hpe), 3- and 14-days post-exposure (dpe). Pools of mosquitoes fed with non-infected blood were also collected for comparisons. Total RNA from each mosquito pool was subjected to RNA-seq analysis and a de novo transcriptome was constructed. A total of 451 differentially expressed genes (DEG) were identified. Most of the transcriptomic alterations were found at an early infection stage after RVFV exposure. Forty-eight DEG related to immune infection-response were characterized. Most of them were related with the RNAi system, Toll and IMD pathways, ubiquitination pathway and apoptosis. Our findings provide for the first time a comprehensive view on Cx. pipiens-RVFV interactions at the molecular level. The early depletion of RNAi pathway genes at the onset of the RVFV infection would allow viral replication in mosquitoes. While genes from the Toll and IMD immune pathways were altered in response to RVFV none of the DEG were related to the JAK/STAT pathway. The fact that most of the DEG involved in the Ubiquitin-proteasome pathway (UPP) or apoptosis were found at an early stage of infection would suggest that apoptosis plays a regulatory role in infected Cx. pipiens midguts. This study provides a number of target genes that could be used to identify new molecular targets for vector control.
Collapse
Affiliation(s)
- Ana I. Núñez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Jèssica Gómez-Garrido
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Marco Brustolin
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sandra Talavera
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Miguel Berdugo
- Instituto de Biología Evolutiva, Universitat Pompeu i Fabra-CSIC, Dr. Aigüader 88, Barcelona, Spain
| | - Marc Dabad
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Tyler Alioto
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
- Universitat Pompeu i Fabra (UPF), Barcelona, Catalonia, Spain
| | - Albert Bensaid
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Núria Busquets
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
34
|
Li HH, Cai Y, Li JC, Su MP, Liu WL, Cheng L, Chou SJ, Yu GY, Wang HD, Chen CH. C-Type Lectins Link Immunological and Reproductive Processes in Aedes aegypti. iScience 2020; 23:101486. [PMID: 32891883 PMCID: PMC7481239 DOI: 10.1016/j.isci.2020.101486] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/14/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
Physiological trade-offs between mosquito immune response and reproductive capability can arise due to insufficient resource availability. C-type lectin family members may be involved in these processes. We established a GCTL-3-/- mutant Aedes aegypti using CRISPR/Cas9 to investigate the role of GCTL-3 in balancing the costs associated with immune responses to arboviral infection and reproduction. GCTL-3-/- mutants showed significantly reduced DENV-2 infection rate and gut commensal microbiota populations, as well as upregulated JAK/STAT, IMD, Toll, and AMPs immunological pathways. Mutants also had significantly shorter lifespans than controls and laid fewer eggs due to defective germ line development. dsRNA knock-down of Attacin and Gambicin, two targets of the AMPs pathway, partially rescued this reduction in reproductive capabilities. Upregulation of immune response following GCTL-3 knock-out therefore comes at a cost to reproductive fitness. Knock-out of other lectins may further improve our knowledge of the molecular and genetic mechanisms underlying reproduction-immunity trade-offs in mosquitoes.
Collapse
Affiliation(s)
- Hsing-Han Li
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, 117604, Singapore; Department of Biological Sciences, National University of Singapore, 117558, Singapore
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Matthew P Su
- Department of Biological Science, Nagoya University, Nagoya 464-8602, Japan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Lie Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Shu-Jen Chou
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Horng-Dar Wang
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan; National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan.
| |
Collapse
|
35
|
MacLeod HJ, Dimopoulos G. Detailed Analyses of Zika Virus Tropism in Culex quinquefasciatus Reveal Systemic Refractoriness. mBio 2020; 11:e01765-20. [PMID: 32817107 PMCID: PMC7439479 DOI: 10.1128/mbio.01765-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 01/01/2023] Open
Abstract
The role of Culex quinquefasciatus in Zika virus transmission has been debated since the epidemic of Zika occurred in the Americas in 2015 to 2016. The majority of studies have found no evidence that C. quinquefasciatus or other Culex species are competent vectors of Zika virus, and the few studies that have proposed Zika vector status for C. quinquefasciatus have relied predominantly on quantitative real-time PCR (qRT-PCR) for viral detection. We assessed the infectious range of pre- and post-epidemic Zika virus isolates in order to classify mosquito samples based on titer infectiousness and demonstrated that two strains of C. quinquefasciatus, including one previously found to be competent, are highly resistant to infection with these Zika isolates compared to Aedes aegypti and are not competent for virus transmission. Further dissection of the dynamics of Zika exposure in both A. aegypti and C. quinquefasciatus revealed that while virus transmission by C. quinquefasciatus is blocked at the levels of the midgut and salivary glands, viral RNA persists in these tissues for prolonged periods post-exposure. We assessed Zika entry dynamics in both Aedes and Culex cells, and our results suggest that Zika virus infection in Culex cells may be blocked downstream of cell entry. These findings strongly suggest that C. quinquefasciatus is not a vector of Zika virus and additionally inform the use of qRT-PCR in vector competence assays as well as our understanding of barriers to arbovirus infection in non-susceptible mosquito species.IMPORTANCE Understanding which mosquito species transmit an emerging arbovirus is critical to effective vector control. During the Zika virus epidemic in 2015 to 2016, Aedes mosquitoes were confirmed as vectors. However, studies addressing the vector status of Culex quinquefasciatus mosquitoes presented conflicting evidence and remain an outstanding source of confusion in the field. Here, we established a robust cell-based assay to identify infectious titers of Zika virus and assessed the virus titers in C. quinquefasciatus by quantitative real-time PCR (qRT-PCR). We found that while low levels of virus were detected in C. quinquefasciatus, these titers did not correspond to infectious virus, and these mosquitoes did not transmit virus in the saliva. We also present evidence that the virus may enter Culex cells before infection is disrupted. Our findings are important for future studies incriminating vector species using qRT-PCR for virus detection and offer new information on how virus transmission is blocked by mosquitoes.
Collapse
Affiliation(s)
- Hannah J MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Rana VS, Popli S, Saurav GK, Yadav K, Kumar A, Sunil S, Kumar N, Singh OP, Natarajan K, Rajagopal R. Aedes aegypti lachesin protein binds to the domain III of envelop protein of Dengue virus-2 and inhibits viral replication. Cell Microbiol 2020; 22:e13200. [PMID: 32141690 DOI: 10.1111/cmi.13200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/31/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022]
Abstract
Dengue virus (DENV) comprises of four serotypes (DENV-1 to -4) and is medically one of the most important arboviruses (arthropod-borne virus). DENV infection is a major human health burden and is transmitted between humans by the insect vector, Aedes aegypti. Ae. aegypti ingests DENV while feeding on infected humans, which traverses through its gut, haemolymph and salivary glands of the mosquito before being injected into a healthy human. During this process of transmission, DENV must interact with many proteins of the insect vector, which are important for its successful transmission. Our study focused on the identification and characterisation of interacting protein partners in Ae. aegypti to DENV. Since domain III (DIII) of envelope protein (E) is exposed on the virion surface and is involved in virus entry into various cells, we performed phage display library screening against domain III of the envelope protein (EDIII) of DENV-2. A peptide sequence showing similarity to lachesin protein was found interacting with EDIII. The lachesin protein was cloned, heterologously expressed, purified and used for in vitro interaction studies. Lachesin protein interacted with EDIII and also with DENV. Further, lachesin protein was localised in neuronal cells of different organs of Ae. aegypti by confocal microscopy. Blocking of lachesin protein in Ae. aegypti with anti-lachesin antibody resulted in a significant reduction in DENV replication.
Collapse
Affiliation(s)
- Vipin S Rana
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Sonam Popli
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo, Toledo, Ohio, USA
| | - Gunjan K Saurav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Department of Zoology, Munshi Lal Arya College, Purnea University, Purnia, Bihar, India
| | - Karuna Yadav
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Delhi, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Delhi, India
| | - Narendra Kumar
- Department of Zoology, Shaheed Mangal Pandey Government Girls Post Graduate College, Meerut, Uttar Pradesh, India
| | - Om P Singh
- National Institute of Malaria Research, Delhi, India
| | | | - Raman Rajagopal
- Gut Biology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
37
|
Zhu Y, Yu X, Cheng G. Insect C-Type Lectins in Microbial Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:129-140. [PMID: 32152945 DOI: 10.1007/978-981-15-1580-4_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
C-type lectins (CTLs) are a family of carbohydrate-recognition domain (CRD)-containing proteins that bind to ligands in a calcium-dependent manner. CTLs act as important components of insect innate immune responses, such as pattern recognition, agglutination, encapsulation, melanization, phagocytosis and prophenoloxidase activation, as well as gut microbiome homeostasis maintenance, to defend against pathogens. Besides, some insect CTLs can facilitate pathogen infection and colonization. In this review, we describe the properties of insect CTLs and focus on explaining their role in viral, bacterial, parasitic and fungal infections.
Collapse
Affiliation(s)
- Yibin Zhu
- Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, 100084, China.,Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China.,School of Life Science, Tsinghua University, Beijing, 100084, China
| | - Xi Yu
- Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, 100084, China.,School of Life Science, Tsinghua University, Beijing, 100084, China
| | - Gong Cheng
- Tsinghua-Peking Joint Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, 100084, China. .,Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
38
|
Sung PS, Hsieh SL. CLEC2 and CLEC5A: Pathogenic Host Factors in Acute Viral Infections. Front Immunol 2019; 10:2867. [PMID: 31867016 PMCID: PMC6909378 DOI: 10.3389/fimmu.2019.02867] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
The protective roles of endosomal toll-like receptors (TLRs) and cytosolic nucleic acid sensors are well elucidated, but the pathogenic host factors during viral infections remain unclear. Spleen tyrosine kinase (Syk)-coupled C-type lectins (CLECs) CLEC2 and CLEC5A are highly expressed on platelets and myeloid cells, respectively. CLEC2 has been shown to recognize snake venom aggretin and the endogenous ligand podoplanin and acts as a critical regulator in the development and immunothrombosis. Although CLEC2 has been reported to interact with type I immunodeficiency virus (HIV-1), its role in viral infections is still unclear. CLEC5A binds to fucose and mannose moieties of dengue virus membrane glycans, as well as to N-acetylglucosamine (GlcNAc)/N-acetylmuramic acid (MurNAc) disaccharides that form the backbone of L. monocytogenes peptidoglycans. Recently, we demonstrated that both CLEC2 and CLEC5A are critical in microbe-induced “neutrophil extracellular trap” (NET) formation and proinflammatory cytokine production. Moreover, activation of CLEC2 by dengue virus (DV) and H5N1 influenza virus (IAV) induces the release of extracellular vesicles (EVs), which further enhance NETosis and proinflammatory cytokine production via CLEC5A and Toll-like receptor 2 (TLR2). These findings not only illustrate the immunomodulatory effects of EVs during platelet-leukocyte interactions, but also demonstrate the critical roles of CLEC2 and CLEC5A in acute viral infections.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
39
|
Pan P, Zhang Q, Liu W, Wang W, Yu Z, Lao Z, Zhang W, Shen M, Wan P, Xiao F, Shereen MA, Zhang W, Tan Q, Liu Y, Liu X, Wu K, Liu Y, Li G, Wu J. Dengue Virus Infection Activates Interleukin-1β to Induce Tissue Injury and Vascular Leakage. Front Microbiol 2019; 10:2637. [PMID: 31824450 PMCID: PMC6883832 DOI: 10.3389/fmicb.2019.02637] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022] Open
Abstract
Dengue virus (DENV) infection causes several diseases ranging from dengue fever to life-threatening dengue hemorrhagic fever and dengue shock syndrome characterized by endothelial dysfunction, vascular leakage, and shock. Here, we identify a potential mechanism by which DENV induces tissue injury and vascular leakage by promoting the activation of interleukin (IL)-1β. DENV facilitates IL-1β secretion in infected patients, mice, human peripheral blood mononuclear cells (PBMCs), mouse bone marrow-derived macrophages (BMDMs), and monocyte-differentiated macrophages (THP-1) via activating the NLRP3 inflammasome. The accumulated data suggest that IL-1β probably induces vascular leakage and tissue injury in interferon-alpha/beta receptor 1 deficient C57BL/6 mice (IFNAR–/– C57BL/6), whereas IL-1 receptor antagonist (IL-1RA) alleviates these effects of IL-1β. Finally, administration of recombinant IL-1β protein results in vascular leakage and tissue injury in C57BL/6 mice. Together, the accumulated results demonstrate that IL-1β contributes to DENV-associated pathology and suggest that IL-1RA acts as a potential agent for the treatment of DENV-associated diseases.
Collapse
Affiliation(s)
- Pan Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Weiyong Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Wenbiao Wang
- Key Laboratory of Virology of Guangzhou, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Zhenyang Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zizhao Lao
- Center for Animal Experiment, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhang
- Center for Animal Experiment, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miaomiao Shen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Pin Wan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Feng Xiao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Muhammad Adnan Shereen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Wen Zhang
- Guangdong LongFan Biological Science and Technology, Foshan, China
| | - Qiuping Tan
- Guangdong LongFan Biological Science and Technology, Foshan, China
| | - Yuntao Liu
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- Center for Animal Experiment, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,Key Laboratory of Virology of Guangzhou, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Geng Li
- Center for Animal Experiment, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,Key Laboratory of Virology of Guangzhou, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
40
|
Ramesh K, Walvekar VA, Wong B, Sayed AMM, Missé D, Kini RM, Mok YK, Pompon J. Increased Mosquito Midgut Infection by Dengue Virus Recruitment of Plasmin Is Blocked by an Endogenous Kazal-type Inhibitor. iScience 2019; 21:564-576. [PMID: 31726374 PMCID: PMC6854080 DOI: 10.1016/j.isci.2019.10.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/20/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Dengue symptoms include alteration of blood coagulation and fibrinolysis, causing severe hemorrhage and death. Here, we demonstrate that higher concentration of plasmin, the human fibrinolytic factor, in blood meal enhances dengue virus (DENV) infection in mosquito midgut and dissemination in mosquitoes. We also show that mosquitoes express a plasmin-selective Kazal-type inhibitor (AaTI) in the midgut to inhibit plasmin proteolysis and revert the enhanced infection. Using bio-layer interferometry, we show that DENV, plasmin, and AaTI interact to form a tripartite complex. Eventually, plasmin increases midgut internalization of dextran molecules and this is reverted by AaTI. Our study demonstrates that (1) DENV recruits plasmin to increase local proteolytic activity in the midgut, thus degrading the glycocalyx and enhancing DENV internalization and (2) AaTI can act as a transmission-blocking agent by inhibiting plasmin proteolysis. Our results indicate that dengue pathogenesis enhances DENV fitness by increasing its infectivity to mosquitoes.
Collapse
Affiliation(s)
- Karthik Ramesh
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Varsha A Walvekar
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Benjamin Wong
- Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Ahmed Mahmoud Mohammed Sayed
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore; Assiut University, Department of Chemistry, Faculty of Science, Assiut 71516, Egypt
| | - Dorothée Missé
- MIVEGEC, UMR IRD 224-CNRS5290-Université de Montpellier, Montpellier, France
| | - R Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore
| | - Yu Keung Mok
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Singapore.
| | - Julien Pompon
- Program in Emerging Infectious Disease, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; MIVEGEC, UMR IRD 224-CNRS5290-Université de Montpellier, Montpellier, France.
| |
Collapse
|
41
|
Yu X, Zhu Y, Xiao X, Wang P, Cheng G. Progress towards Understanding the Mosquito-Borne Virus Life Cycle. Trends Parasitol 2019; 35:1009-1017. [PMID: 31669148 DOI: 10.1016/j.pt.2019.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/02/2019] [Accepted: 09/16/2019] [Indexed: 12/30/2022]
Abstract
Mosquito-borne arboviruses are a group of heterogeneous viruses that are mainly transmitted to vertebrate hosts and are the aetiological agents of many human diseases. These viruses naturally maintain a life cycle between distinct hosts by transmission from an infected mosquito to a naive host, and acquisition from a viraemic host back to a fed mosquito. To survive in and maintain a cycle between different host environments, mosquito-borne arboviruses exploit sophisticated approaches, including subverting the immune system, hijacking host factors, and taking advantage of gut microbes. We summarize the recent progress towards understanding the mechanisms of arboviral transmission and acquisition by mosquitoes. This knowledge offers an insight into the emergence and re-emergence of arboviruses in nature and an avenue for disease prevention in the future.
Collapse
Affiliation(s)
- Xi Yu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China; School of Life Sciences, 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
| | - Xiaoping Xiao
- 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
| | - Penghua Wang
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, CT 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
|
42
|
Watanabe Y, Bowden TA, Wilson IA, Crispin M. Exploitation of glycosylation in enveloped virus pathobiology. Biochim Biophys Acta Gen Subj 2019; 1863:1480-1497. [PMID: 31121217 PMCID: PMC6686077 DOI: 10.1016/j.bbagen.2019.05.012] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022]
Abstract
Glycosylation is a ubiquitous post-translational modification responsible for a multitude of crucial biological roles. As obligate parasites, viruses exploit host-cell machinery to glycosylate their own proteins during replication. Viral envelope proteins from a variety of human pathogens including HIV-1, influenza virus, Lassa virus, SARS, Zika virus, dengue virus, and Ebola virus have evolved to be extensively glycosylated. These host-cell derived glycans facilitate diverse structural and functional roles during the viral life-cycle, ranging from immune evasion by glycan shielding to enhancement of immune cell infection. In this review, we highlight the imperative and auxiliary roles glycans play, and how specific oligosaccharide structures facilitate these functions during viral pathogenesis. We discuss the growing efforts to exploit viral glycobiology in the development of anti-viral vaccines and therapies.
Collapse
Affiliation(s)
- Yasunori Watanabe
- School of Biological Sciences and Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK; Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK; Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Thomas A Bowden
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Oxford OX3 7BN, UK
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Max Crispin
- School of Biological Sciences and Institute of Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
43
|
Caragata EP, Tikhe CV, Dimopoulos G. Curious entanglements: interactions between mosquitoes, their microbiota, and arboviruses. Curr Opin Virol 2019; 37:26-36. [PMID: 31176069 PMCID: PMC6768729 DOI: 10.1016/j.coviro.2019.05.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 11/22/2022]
Abstract
Mosquitoes naturally harbor a diverse community of microorganisms that play a crucial role in their biology. Mosquito-microbiota interactions are abundant and complex. They can dramatically alter the mosquito immune response, and impede or enhance a mosquito's ability to transmit medically important arboviral pathogens. Yet critically, given the massive public health impact of arboviral disease, few such interactions have been well characterized. In this review, we describe the current state of knowledge of the role of microorganisms in mosquito biology, how microbial-induced changes to mosquito immunity moderate infection with arboviruses, cases of mosquito-microbial-virus interactions with a defined mechanism, and the molecular interactions that underlie the endosymbiotic bacterium Wolbachia's ability to block virus infection in mosquitoes.
Collapse
Affiliation(s)
- Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Chinmay V Tikhe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
44
|
Campos RK, Garcia-Blanco MA, Bradrick SS. Roles of Pro-viral Host Factors in Mosquito-Borne Flavivirus Infections. Curr Top Microbiol Immunol 2019; 419:43-67. [PMID: 28688087 DOI: 10.1007/82_2017_26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Identification and analysis of viral host factors is a growing area of research which aims to understand the how viruses molecularly interface with the host cell. Investigations into flavivirus-host interactions has led to new discoveries in viral and cell biology, and will potentially bolster strategies to control the important diseases caused by these pathogens. Here, we address the current knowledge of prominent host factors required for the flavivirus life-cycle and mechanisms by which they promote infection.
Collapse
Affiliation(s)
- Rafael K Campos
- Department of Molecular Genetics and Microbiology, Center for RNA Biology, Duke University, Durham, NC, USA.,Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA. .,Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
45
|
Meng Q, Zhang J, Zhang H, Zhou G, Ni R, Zhao Y, Qin Q, Zou Z. Comparative analysis of C-type lectin domain proteins in the ghost moth, Thitarodes xiaojinensis (Lepidoptera: Hepialidae). INSECT SCIENCE 2019; 26:453-465. [PMID: 29274206 PMCID: PMC7379682 DOI: 10.1111/1744-7917.12564] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/03/2017] [Accepted: 11/19/2017] [Indexed: 05/24/2023]
Abstract
Insects have a large family of C-type lectins involved in cell adhesion, pathogen recognition and activation of immune responses. In this study, 32 transcripts encoding C-type lectin domain proteins (CTLDPs) were identified from the Thitarodes xiaojinensis transcriptome. According to their domain structures, six CTLDPs with one carbohydrate-recognition domain (CRD) were classified into the CTL-S subfamily. The other 23 CTLDPs with two CRDs were grouped into the immulectin (IML) subfamily. The remaining three with extra regulatory domains were sorted into the CTL-X subfamily. Phylogenetic analysis showed that CTL-S and CTL-X members from different insects could form orthologous groups. In contrast, no T. xiaojinensis IML orthologues were found in other insects. Remarkable lineage-specific expansion in this subfamily was observed reflecting that these CTLDPs, as important receptors, have evolved diversified members in response to a variety of microbes. Prediction of binding ligands revealed that T. xiaojinensis, a cold-adapted species, conserved the ability of CRDs to combine with Ca2+ to keep its receptors from freezing. Comparative analysis of induction of CTLDP genes after different immune challenges indicated that IMLs might play critical roles in immune defenses. This study examined T. xiaojinensis CTLDPs and provides a basis for further studies of their characteristics.
Collapse
Affiliation(s)
- Qian Meng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Ji‐Hong Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Huan Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Gui‐Ling Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Ruo‐Yao Ni
- College of Life SciencesHebei UniversityBaodingHebeiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yan‐Ni Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qi‐Lian Qin
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of ZoologyChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
46
|
Hu D, Zhu Z, Li S, Deng Y, Wu Y, Zhang N, Puri V, Wang C, Zou P, Lei C, Tian X, Wang Y, Zhao Q, Li W, Prabakaran P, Feng Y, Cardosa J, Qin C, Zhou X, Dimitrov DS, Ying T. A broadly neutralizing germline-like human monoclonal antibody against dengue virus envelope domain III. PLoS Pathog 2019; 15:e1007836. [PMID: 31242272 PMCID: PMC6615639 DOI: 10.1371/journal.ppat.1007836] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 07/09/2019] [Accepted: 05/13/2019] [Indexed: 11/19/2022] Open
Abstract
Dengue is the most widespread vector-borne viral disease caused by dengue virus (DENV) for which there are no safe, effective drugs approved for clinical use. Here, by using sequential antigen panning of a yeast antibody library derived from healthy donors against the DENV envelop protein domain III (DIII) combined with depletion by an entry defective DIII mutant, we identified a cross-reactive human monoclonal antibody (mAb), m366.6, which bound with high affinity to DENV DIII from all four DENV serotypes. Immunogenetic analysis indicated that m366.6 is a germline-like mAb with very few somatic mutations from the closest VH and Vλ germline genes. Importantly, we demonstrated that it potently neutralized DENV both in vitro and in the mouse models of DENV infection without detectable antibody-dependent enhancement (ADE) effect. The epitope of m366.6 was mapped to the highly conserved regions on DIII, which may guide the design of effective dengue vaccine immunogens. Furthermore, as the first germline-like mAb derived from a naïve antibody library that could neutralize all four DENV serotypes, the m366.6 can be a tool for exploring mechanisms of DENV infection, and is a promising therapeutic candidate.
Collapse
Affiliation(s)
- Dan Hu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongyu Zhu
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Shun Li
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Fudan University, Shanghai, China
| | - Yongqiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yanling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Nana Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Vinita Puri
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Chunyu Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Fudan University, Shanghai, China
| | - Cheng Lei
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaolong Tian
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yulu Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Zhao
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Wei Li
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Ponraj Prabakaran
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Yang Feng
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Jane Cardosa
- Institute of Health and Community Medicine, Universiti Malaysia Sarawak, Malaysia
| | - Chengfeng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Fudan University, Shanghai, China
| | - Dimiter S. Dimitrov
- National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Tianlei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Aedes aegypti HPX8C modulates immune responses against viral infection. PLoS Negl Trop Dis 2019; 13:e0007287. [PMID: 30986216 PMCID: PMC6464178 DOI: 10.1371/journal.pntd.0007287] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
Mosquitoes act as vectors of numerous pathogens that cause human diseases. Dengue virus (DENV) transmitted by mosquito, Aedes aegypti, is responsible for dengue fever epidemics worldwide with a serious impact on human health. Currently, disease control mainly relies on vector targeted intervention strategies. Therefore, it is imperative to understand the molecular mechanisms underlying the innate immune response of mosquitoes against pathogens. In the present study, the expression profiles of immunity-related genes in the midgut responding to DENV infection by feeding were analyzed by transcriptome and quantitative real-time PCR. The level of Antimicrobial peptides (AMPs) increased seven days post-infection (d.p.i.), which could be induced by the Toll immune pathway. The expression of reactive oxygen species (ROS) genes, including antioxidant genes, such as HPX7, HPX8A, HPX8B, HPX8C were induced at one d.p.i. and peaked again at ten d.p.i. in the midgut. Interestingly, down-regulation of the antioxidant gene HPX8C by RNA interference led to reduction in the virus titer in the mosquito, probably due to the elevated levels of ROS. Application of a ROS inhibitor and scavenger molecules further established the role of oxygen free radicals in the modulation of the immune response to DENV infection. Overall, our comparative transcriptome analyses provide valuable information about the regulation of immunity related genes in the transmission vector in response to DENV infection. It further allows us to identify novel molecular mechanisms underlying the host-virus interaction, which might aid in the development of novel strategies to control mosquito-borne diseases. HPX8C is a heme-containing peroxidase, which can move reactive oxygen species (ROS) damage to the organism by reducing H2O2 to H2O. Previously, the peroxidase gene has been shown to modulate midgut immunity and regulate anti-malarial response in mosquitoes. In this study, the classical immune signaling pathways, Toll and IMD genes might be late responses against the viruses. HPX8C was demonstrated here to play a role in antiviral immunity against DENV infection in Ae. Aegypti mosquitoes. HPX8C expression was induced by DENV infection and continued to increase with an elevated virus titer. In HPX8C-depleted mosquitoes, the ROS level was found to be increased with a corresponding decrease in the DENV and ZIKV virus titer. Therefore, it was speculated that HPX8C mediated immune responses against the DENV in the mosquito in the late stage of viral infection, which could be controlled by Toll pathway.
Collapse
|
48
|
Adaptive genetic diversifications among tick-borne encephalitis virus subtypes: A genome-wide perspective. Virology 2019; 530:32-38. [DOI: 10.1016/j.virol.2019.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 12/30/2022]
|
49
|
Abstract
Mosquitoes are haematophagous vectors for hundreds of pathogenic viruses that are aetiological agents of human diseases. In nature, mosquito-borne viruses maintain a lifecycle between mosquitoes and vertebrate animals. Viruses are acquired by a naive mosquito from an infected host by blood meals and then propagate extensively in the mosquito's tissues. This mosquito then becomes a virus reservoir and is competent to transmit the viruses to a naive vertebrate host through the next blood meal. To survive in and efficiently cycle between two distinct host environments, mosquito-borne viruses have evolved delicate and smart strategies to comprehensively exploit host and vector factors. Here, we provide an update on recent studies of the mechanisms of virus survival in, acquisition and transmission by mosquitoes.
Collapse
|
50
|
Early Events in Japanese Encephalitis Virus Infection: Viral Entry. Pathogens 2018; 7:pathogens7030068. [PMID: 30104482 PMCID: PMC6161159 DOI: 10.3390/pathogens7030068] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne zoonotic flavivirus, is an enveloped positive-strand RNA virus that can cause a spectrum of clinical manifestations, ranging from mild febrile illness to severe neuroinvasive disease. Today, several killed and live vaccines are available in different parts of the globe for use in humans to prevent JEV-induced diseases, yet no antivirals are available to treat JEV-associated diseases. Despite the progress made in vaccine research and development, JEV is still a major public health problem in southern, eastern, and southeastern Asia, as well as northern Oceania, with the potential to become an emerging global pathogen. In viral replication, the entry of JEV into the cell is the first step in a cascade of complex interactions between the virus and target cells that is required for the initiation, dissemination, and maintenance of infection. Because this step determines cell/tissue tropism and pathogenesis, it is a promising target for antiviral therapy. JEV entry is mediated by the viral glycoprotein E, which binds virions to the cell surface (attachment), delivers them to endosomes (endocytosis), and catalyzes the fusion between the viral and endosomal membranes (membrane fusion), followed by the release of the viral genome into the cytoplasm (uncoating). In this multistep process, a collection of host factors are involved. In this review, we summarize the current knowledge on the viral and cellular components involved in JEV entry into host cells, with an emphasis on the initial virus-host cell interactions on the cell surface.
Collapse
|