1
|
Lima RLS, Menegatto MBDS, Almeida LT, Magalhães JCD, Ferraz AC, Magalhães CLDB. Silymarin exerts antioxidant and antiviral effects on Zika virus infection. J Virol Methods 2025; 335:115133. [PMID: 40043812 DOI: 10.1016/j.jviromet.2025.115133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
The Zika virus (ZIKV) - Orthoflavivirus zikaense - epidemic and its association with severe neurological disorders have created an urgent need to understand the disease pathogenesis and identify potential therapeutic targets. In previous investigations, we have shown that oxidative stress is associated with the pathogenesis of ZIKV infection in vitro and in vivo, and that silymarin has anti-ZIKV action in vitro. Here, we characterised the antioxidant and antiviral effects of silymarin against ZIKV infection in an animal model. We observed an increase in the levels of biomarkers of oxidative damage and in antioxidant enzyme activities in the livers of ZIKV-infected C57BL/6 mice. However, these effects were reversed in ZIKV-infected animals that were treated with silymarin. Furthermore, silymarin reduced the viral load in the livers of animals. Next, by in vitro studies, we confirmed that the anti-ZIKV action of silymarin is independent of its antioxidant activity. This work reinforces the potential use of silymarin against Zika fever.
Collapse
Affiliation(s)
- Rafaela Lameira Souza Lima
- Programa de Pós Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Marília Bueno da Silva Menegatto
- Programa de Pós Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | | | - José Carlos de Magalhães
- Departamento de Química, Biotecnologia e Engenharia de Bioprocessos, Universidade Federal de São João del-Rei, Ouro Branco, Minas Gerais, Brazil
| | - Ariane Coelho Ferraz
- Programa de Pós Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| | - Cintia Lopes de Brito Magalhães
- Programa de Pós Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Programa de Pós Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, NUPEB, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
2
|
Lai M, Lai R, He B, Wang X, Chen L, Mo Q. Robust antiviral innate immune response and miRNA regulatory network were identified in ZIKV-infected cells: implications in the pathogenesis of ZIKV infection. Virus Genes 2025; 61:249-264. [PMID: 39955676 DOI: 10.1007/s11262-025-02136-4] [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: 12/14/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Zika virus (ZIKV) infection has emerged as a significant public health concern due to its association with fetal microcephaly and Guillain-Barre syndrome (GBS). Unfortunately, its detailed pathogenesis remains unclear. To better understand how ZIKV evades host antiviral immunity, we analyzed the microarray dataset (GSE98889) of ZIKV-infected primary human brain microvascular endothelial cells (hBMECs) retrieved from the gene expression omnibus (GEO). 160, 1423, 969, 829, and 600 differentially expressed genes (DEGs) were identified at 12, 24, 48, 72, and 216 hours post-ZIKV infection in hBMECs, respectively. Subsequently, 31 common DEGs across all time-points were selected for further analysis. Gene ontology (GO) functional analysis showed these 31 DEGs were mainly involved in the host antiviral innate immune responses. Protein-protein interaction (PPI) network analysis identified 10 hub genes (MX1, OAS1, OAS2, IFI44, IFI44L, IFIT1, IFIT2, IFIT3, IFIH1, and XAF1), which were all interferon-stimulated genes (ISGs) and upregulated. qRT-PCR was used to validate the expression patterns of these 10 hub genes in different ZIKV-infected cell lines. Finally, miRNA-mRNA regulatory network analysis revealed that hsa-miR-129-2-3p, hsa-miR-138-5p, hsa-miR-21-3p, hsa-miR-27a-5p, hsa-miR-449a, and hsa-miR449b-5p were key miRNAs regulating these hub genes. Our study showed that ZIKV infection activated the host innate immune response to restrict ZIKV infection. The common pathways, hub genes, and their regulatory miRNA network offer new insights into virus-host interactions, enhancing our understanding of ZIKV pathogenesis.
Collapse
Affiliation(s)
- Mingshuang Lai
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Rongji Lai
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Baoren He
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Xinwei Wang
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Limin Chen
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Qiuhong Mo
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China.
| |
Collapse
|
3
|
Siew ZY, Seow I, Lim XR, Tang CZ, Djamil FM, Ong GK, Leong PP, Wong ST, Voon K. Arboviruses: the hidden danger of the tropics. Arch Virol 2025; 170:140. [PMID: 40418376 DOI: 10.1007/s00705-025-06314-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/17/2025] [Indexed: 05/27/2025]
Abstract
Arboviruses are viruses that are transmitted by arthropods such as mosquitoes, ticks, and flies, and most of them are RNA viruses. Vector-borne transmission occurs when an infected arthropod bites a vertebrate host, allowing the virus to enter the bloodstream and initiate infection. Arboviruses are known to cause significant morbidity and mortality in mammals, and at least a hundred of them have been identified as human pathogens. In this review, we provide an updated overview of four prominent arboviruses that are present in Southeast Asia (SEA): dengue virus (DENV), Japanese encephalitis virus (JEV), Zika virus (ZIKV), and chikungunya virus (CHIKV). The epidemiology and pathogenesis of these viruses and the currently used methods for diagnosis, prevention, and treatment of arbovirus infections are discussed in detail. Finally, we summarise the concerns and future considerations for combating these dangerous pathogens.
Collapse
Affiliation(s)
- Zhen Yun Siew
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia.
| | - Isaac Seow
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Xin Rui Lim
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Chen Zhe Tang
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia
| | - Fadhilah Moh Djamil
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ghee Khang Ong
- School of Medicine, IMU University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Pooi Pooi Leong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, 43000, Kajang, Malaysia
| | - Siew Tung Wong
- Department of Pathology and Pharmacology, School of Medicine, IMU University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Kenny Voon
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
4
|
Fan JJ, Hu C, Hu M, Dong WS, Li K, Ye YJ, Zhang X. A brief overview of the E3 ubiquitin ligase: TRIM7. Cell Signal 2025:111886. [PMID: 40419231 DOI: 10.1016/j.cellsig.2025.111886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025]
Abstract
TRIM7, a member of the E3 ubiquitin ligase family, has garnered significant attentions in different research fields since its discovery. This enzyme plays indispensable roles in various pathophysiological processes through ubiquitination-mediated degradation of diverse protein substrates. This review systematically summarizes the current knowledge on the protein structure and biological functions of TRIM7. Structurally, TRIM7 features a conserved RBCC motif (RING, B-box, and coiled-coil domains) coupled with a variable C-terminal region that dictates the substrate specificity. In infectious contexts, TRIM7 is required for the pathogen-specific regulation, and exerts paradoxical effects by either promoting host defense or facilitating viral pathogenesis depending on pathogen type. Within oncology, TRIM7 manifests tumor-suppressive properties through regulating metastasis, apoptosis, and tumor immunology. In addition, it might serve as a reliable biomarker for monitoring the progression of idiopathic pulmonary fibrosis and also inhibits the progression of atherosclerosis. In summary, TRIM7 plays critical roles in different pathophysiological processes, and it might be a predictive and therapeutic target in certain human diseases.
Collapse
Affiliation(s)
- Jun-Jie Fan
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Can Hu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Kang Li
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xin Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China.
| |
Collapse
|
5
|
Wiemers P, Graf I, Addo MM, Arck PC, Diemert A. Mothers and mosquitoes: climate change contributes to the spread of vector-borne pathogens posing a substantial threat to pregnant women. Semin Immunopathol 2025; 47:25. [PMID: 40272573 PMCID: PMC12021716 DOI: 10.1007/s00281-025-01050-z] [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: 08/07/2024] [Accepted: 03/23/2025] [Indexed: 04/25/2025]
Abstract
Infectious diseases have threatened individuals and societies since the dawn of humanity. Certain population groups, including pregnant women, young children and the elderly, are particularly vulnerable to severe infections. Over the past few centuries, advances in medical standards and the availability of vaccines have reduced infection-related mortality and morbidity rates in industrialized countries. However, the global rise in temperatures and increased precipitation present a new challenge, facilitating the broader distribution of disease vectors, such as mosquitoes, bugs and ticks, to higher altitudes and latitudes. Consequently, epidemic and pandemic outbreaks associated with these vectors, such as Zika, West Nile, dengue, yellow fever, chikungunya and malaria, are increasingly impacting diverse populations. This review comprehensively examines how infections associated with climate change disproportionately affect the health and well-being of pregnant women and their unborn children. There has been a noticeable emergence of vector-borne diseases in Europe. Consequently, we stress the importance of implementing measures that effectively protect pregnant women from these increasing infections globally and regionally. We advocate for initiatives to safeguard pregnant women from these emerging threats, beginning with enhanced education to raise awareness about the evolving risks this particularly vulnerable population faces.
Collapse
Affiliation(s)
- Pauline Wiemers
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Graf
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Petra C Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Center for Child and Adolescent Health, Partner Site Hamburg, Hamburg, Germany.
| | - Anke Diemert
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Center for Child and Adolescent Health, Partner Site Hamburg, Hamburg, Germany.
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany.
| |
Collapse
|
6
|
Yu W, Tao J, Cao H, Zheng W, Zhang B, Zhang Y, Xu P, Zhang Y, Liu X, Wang Y, Cai H, Liu G, Liu F, Wang H, Zhao H, Mysorekar IU, Hu X, Cao B. The HAVCR1-centric host factor network drives Zika virus vertical transmission. Cell Rep 2025; 44:115464. [PMID: 40156834 DOI: 10.1016/j.celrep.2025.115464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/11/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
Zika virus (ZIKV) vertical transmission results in devastating congenital malformations and pregnancy complications; however, the specific receptor and host factors facilitating ZIKV maternal-fetal transmission remain elusive. Here, we employ a genome-wide CRISPR screening and identify multiple placenta-intrinsic factors modulating ZIKV infection. Our study unveils that hepatitis A virus cellular receptor 1 (HAVCR1) serves as a primary receptor governing ZIKV entry in placental trophoblasts. The GATA3-HAVCR1 axis regulates heterogeneous cell tropism in the placenta. Notably, placenta-specific Havcr1 deletion in mice significantly impairs ZIKV transplacental transmission and associated adverse pregnancy outcomes. Mechanistically, the immunoglobulin variable-like domain of HAVCR1 binds to ZIKV via domain III of envelope protein and virion-associated phosphatidylserine. Proteomic profiling and function analyses reveal that AP2S1 cooperates with HAVCR1 for ZIKV internalization through clathrin-mediated endocytosis. Overall, our work underscores the pivotal role of HAVCR1 in mediating ZIKV vertical transmission and highlights a therapeutic target for alleviating congenital Zika syndrome.
Collapse
Affiliation(s)
- Wenzhe Yu
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Jun Tao
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Hongmin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Wanshan Zheng
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Beiang Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yue Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Peiqun Xu
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yiwei Zhang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Xuan Liu
- State Key Laboratory of Vaccine for Infectious Disease, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Gang Liu
- State Key Laboratory of Vaccine for Infectious Disease, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China
| | - Fan Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Haiyan Zhao
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, Wuhan University, Wuhan 430072, Hubei, China
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoqian Hu
- State Key Laboratory of Vaccine for Infectious Disease, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen 361102, Fujian, China.
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China; Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
7
|
Wang X, Qian C, Zhang C, Hu S, Asad M, Yang C, Liao B, Guo X, Zhang C, Li Q, Li X, Huang Q, Si Y, Zhu B, Cao S, Ye J. Zika virus transmission in Aedes aegypti: A systematic study on the ability of mosquitoes to transmit the virus horizontally and vertically. Virol Sin 2025; 40:192-205. [PMID: 39947399 PMCID: PMC12131025 DOI: 10.1016/j.virs.2025.02.001] [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: 07/13/2024] [Accepted: 02/07/2025] [Indexed: 04/08/2025] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus belonging to the genus Orthoflavivirus, and the family Flaviviridae. It commonly presents with febrile-like symptoms, neurological issues, and pregnancy complications in humans. Currently, there is no commercial vaccine or specific treatment available to prevent ZIKV infection. Therefore, controlling the epidemic's spread relies on preventing mosquitoes from transmitting the virus. Although various studies have explored the transmission of ZIKV between mosquitoes and vertebrate hosts, comprehensive research on potential mosquito-to-mosquito transmission of ZIKV remains limited. In this study, we conducted systematic laboratory investigations to assess the ability of ZIKV to spread among mosquitoes, and to evaluate the impact of ZIKV infection on mosquito development. Our findings revealed that ZIKV can be transmitted between Aedes aegypti mosquitoes both vertically and horizontally, through oviposition and contact between mosquitoes of the same or opposite sex. Additionally, we observed that ZIKV infection resulted in a reduction in the number of mosquito eggs but an increase in their size. The widespread distribution of ZIKV in infected mosquitoes and the altered levels of hormone related genes following viral infection were noted, which may contribute to viral transmission among mosquitoes and affect mosquito development. This research provides systematic experimental evidence of ZIKV transmission among mosquitoes, which is crucial for developing novel strategies to disrupt the spread of orthoflaviviruses and other mosquito-borne pathogens.
Collapse
Affiliation(s)
- Xugang Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chaonan Qian
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chenxi Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Siyun Hu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Muhammad Asad
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chengjie Yang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bingrui Liao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaotong Guo
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chen Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Quanzhi Li
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xinyao Li
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Qiuying Huang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
| | - Youhui Si
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bibo Zhu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shengbo Cao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jing Ye
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
8
|
Pereira CADM, Mendes RPG, da Silva PG, Chaves EJF, Pena LJ. Vaccines Against Urban Epidemic Arboviruses: The State of the Art. Viruses 2025; 17:382. [PMID: 40143310 PMCID: PMC11945797 DOI: 10.3390/v17030382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Arboviruses represent a contemporary global challenge, prompting coordinated efforts from health organizations and governments worldwide. Dengue, chikungunya, and Zika viruses have become endemic in the tropics, resulting in the so-called "triple arbovirus epidemic". These viruses are transmitted typically through the bites of infected mosquitoes, especially A. aegypti and A. albopictus. These mosquito species are distributed across all continents and exhibit a high adaptive capacity in diverse environments. When combined with unplanned urbanization, uncontrolled population growth, and international travel-the so-called "triad of the modern world"-the maintenance and spread of these pathogens to new areas are favored. This review provides updated information on vaccine candidates targeting dengue, chikungunya, and Zika viruses. Additionally, we discuss the challenges, perspectives, and issues associated with their successful production, testing, and deployment within the context of public health.
Collapse
Affiliation(s)
| | | | | | | | - Lindomar José Pena
- Laboratory of Virology and Experimental Therapy (Lavite), Department of Virology, Aggeu Magalhães Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), 50670-420 Recife, Brazil; (C.A.d.M.P.); (R.P.G.M.); (P.G.d.S.); (E.J.F.C.)
| |
Collapse
|
9
|
Saeed H, Rehman G, Mehmood Qadri H, Sohail A, Ul Haq A, Sadiq HZ, Yasin S, Khalid Rana MA. Neurological Manifestations of Zika Virus Infection: An Updated Review of the Existing Literature. Cureus 2025; 17:e80960. [PMID: 40260336 PMCID: PMC12010016 DOI: 10.7759/cureus.80960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
Zika virus (ZIKV) is a neurotropic virus closely linked to other flaviviruses like dengue virus, West Nile virus, yellow fever, and Japanese encephalitis virus. Though initially considered a mild virus, ZIKV gained everybody's attention when the World Health Organization (WHO) declared it a global public health emergency in February 2016. Being considered an important cause of innumerable neurological manifestations and pediatric modality, we aimed to present a comprehensive overview of the neurological details of ZIKV infection. This study reviews the neurological manifestations of ZIKV infection. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) strategy was employed, along with a combination of keywords, to enlist all articles with data on ZIKV and its neurological manifestations, diagnosis, and treatment. All case reports, case series, and systematic reviews published between 2017 and 2024, focusing on neurological manifestations of ZIKV, were included in this study. Case studies, editorials, letters to the editors, and clinical images were excluded. The search was conducted using Boolean operators "AND" and "OR" on PubMed and Google Scholar. A total of five case reports, one case series, and one systematic review and meta-analysis were included. Out of 603 patients, the study suggested a male preponderance of 366 patients (62.5%) for ZIKV infection. About 258 patients presented with rash (46.1%), 243 with fever (43.8%), and 134 with dysphagia (36.5%). Neurological signs on examination were limb paresis in 545 (91.1%) patients, areflexia in 401 (88.9%) patients, and tetraparesis in 153 (61%) patients. A significant finding on magnetic resonance imaging (MRI) showed enhancement of the distal cord, conus medullaris, and cauda equina in two cases (0.3%). Serological analysis showed a positive plaque reduction neutralization test (PRNT) in 125 (73.5%) patients. Increased protein levels were identified in 240 (78.7%) cases on cerebrospinal fluid (CSF) analysis. The commonest diagnostic modality utilized was polymerase chain reaction (PCR) in 118 (24.3%) cases. Intravenous immunoglobulins (IVIg) were used for the medical management of 442 patients included in this review (77.4%). ZIKV is known to cause insidious detrimental effects on the central nervous system regardless of the age of an individual. Being a cause of extreme sensorimotor disability, various preventive and precautionary measures are being undertaken to ensure early diagnosis and prevent prolonged liability on a patient's health. Effective therapeutics including IVIg have paved the way in bringing down the hurdles in the management and cure of the infection.
Collapse
Affiliation(s)
- Hasan Saeed
- Pathology, Shifa International Hospital, Islamabad, PAK
| | - Gohar Rehman
- Internal Medicine, Allama Iqbal Medical College, Lahore, PAK
| | - Haseeb Mehmood Qadri
- General Surgery, Lahore General Hospital, Lahore, PAK
- Neurological Surgery, Punjab Institute of Neurosciences, Lahore, PAK
| | - Amna Sohail
- Internal Medicine, Lahore General Hospital, Lahore, PAK
| | - Arshaman Ul Haq
- Internal Medicine, Aziz Bhatti Shaheed Teaching Hospital, Gujrat, PAK
| | | | - Shahnila Yasin
- Internal Medicine, Nawaz Sharif Medical College, Gujrat, PAK
| | | |
Collapse
|
10
|
dos Santos ALS, Rosolen BB, Ferreira FC, Chiancone IS, Pereira SS, Pontes KFM, Traina E, Werner H, Granese R, Araujo Júnior E. Intrauterine Zika Virus Infection: An Overview of the Current Findings. J Pers Med 2025; 15:98. [PMID: 40137414 PMCID: PMC11943202 DOI: 10.3390/jpm15030098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of the family Flaviviridae. The association between ZIKV and microcephaly was first described in Brazil in 2015. The risk of vertical transmission occurs in pregnant women with or without symptoms, and the risk of malformation appears to be worse when infection occurs in the first and second trimesters of pregnancy. The rate of vertical transmission varies from 26 to 65%, and not all fetuses develop malformations. The incidence of malformations resulting from transmission is uncertain, ranging from 6-8% in the US to 40% in Brazil. Congenital ZIKV syndrome is a set of clinical manifestations that can affect the fetus of a mother infected with ZIKV. The manifestations are broad and nonspecific, including microcephaly, subcortical calcifications, ocular changes, congenital contractures, early hypertension, and pyramidal and extrapyramidal signs. Other findings such as growth restriction and fetal miscarriage/death may also occur. Our aim in this article is to review the literature on mosquito transmission, clinical presentation, serologic diagnosis, intrauterine transmission, pre- and postnatal imaging diagnostic findings, and short- and long-term follow-up.
Collapse
Affiliation(s)
- Ana Luiza Soares dos Santos
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Beatriz Bussi Rosolen
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Fernanda Curvelo Ferreira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Isabella Samões Chiancone
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Stefany Silva Pereira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Karina Felippe Monezi Pontes
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
- Service of Gynecology and Obstetrics, Ipiranga Hospital, São Paulo 04262-000, SP, Brazil
| | - Evelyn Traina
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| | - Heron Werner
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro 22453-900, RJ, Brazil;
| | - Roberta Granese
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, “G. Martino” University Hospital, 98100 Messina, Italy
| | - Edward Araujo Júnior
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| |
Collapse
|
11
|
Lackritz EM, Ng LC, Marques ETA, Rabe IB, Bourne N, Staples JE, Méndez-Rico JA, Harris E, Brault AC, Ko AI, Beasley DWC, Leighton T, Wilder-Smith A, Ostrowsky JT, Mehr AJ, Ulrich AK, Velayudhan R, Golding JP, Fay PC, Cehovin A, Moua NM, Moore KA, Osterholm MT, Barrett ADT. Zika virus: advancing a priority research agenda for preparedness and response. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00794-1. [PMID: 40024263 DOI: 10.1016/s1473-3099(24)00794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 03/04/2025]
Abstract
The 2015-16 Zika virus epidemic emerged in the Americas and rapidly spread throughout the region and beyond, showing the epidemic potential of this mosquito-borne Orthoflavivirus and its capacity to cause severe congenital malformations and neurological sequelae. WHO declared the Zika virus epidemic a public health emergency of international concern in 2016. Despite this declaration, there are no licensed Zika virus vaccines, therapeutics, or diagnostic tests appropriate for routine antenatal screening. To address this absence of essential tools to detect and mitigate the threat of future Zika virus outbreaks, a group of global experts developed a priority agenda for Zika virus research and development. This Series paper summarises crucial challenges and knowledge gaps and outlines a comprehensive strategy to advance research, surveillance, global capacity, policy, and investment for Zika virus preparedness and response.
Collapse
Affiliation(s)
- Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA.
| | - Lee-Ching Ng
- National Environment Agency, Environmental Health Institute, Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Ernesto T A Marques
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Virology and Experimental Therapeutics, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
| | | | - Nigel Bourne
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - J Erin Staples
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | - Jairo A Méndez-Rico
- Pan American Health Organization, WHO Region of the Americas, Washington, DC, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Aaron C Brault
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | - Albert I Ko
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA; Ministry of Health, Salvador, Brazil
| | - David W C Beasley
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | | | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Petra C Fay
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | - Ana Cehovin
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | - Nicolina M Moua
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
12
|
Devaraj AR, Marianthiran VJ. Advancements in Viral Genomics: Gated Recurrent Unit Modeling of SARS-CoV-2, SARS, MERS, and Ebola viruses. Rev Soc Bras Med Trop 2025; 58:e004012024. [PMID: 39936709 PMCID: PMC11805527 DOI: 10.1590/0037-8682-0178-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/08/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Emerging infections have posed persistent threats to humanity throughout history. Rapid and unprecedented anthropogenic, behavioral, and social transformations witnessed in the past century have expedited the emergence of novel pathogens, intensifying their impact on the global human population. METHODS This study aimed to comprehensively analyze and compare the genomic sequences of four distinct viruses: SARS-CoV-2, SARS, MERS, and Ebola. Advanced genomic sequencing techniques and a Gated Recurrent Unit-based deep learning model were used to examine the intricate genetic makeup of these viruses. The proposed study sheds light on their evolutionary dynamics, transmission patterns, and pathogenicity and contributes to the development of effective diagnostic and therapeutic interventions. RESULTS This model exhibited exceptional performance as evidenced by accuracy values of 99.01%, 98.91%, 98.35%, and 98.04% for SARS-CoV-2, SARS, MERS, and Ebola respectively. Precision values ranged from 98.1% to 98.72%, recall values consistently surpassed 92%, and F1 scores ranged from 95.47% to 96.37%. CONCLUSIONS These results underscore the robustness of this model and its potential utility in genomic analysis, paving the way for enhanced understanding, preparedness, and response to emerging viral threats. In the future, this research will focus on creating better diagnostic instruments for the early identification of viral illnesses, developing vaccinations, and tailoring treatments based on the genetic composition and evolutionary patterns of different viruses. This model can be modified to examine a more extensive variety of diseases and recently discovered viruses to predict future outbreaks and their effects on global health.
Collapse
Affiliation(s)
- Abhishak Raj Devaraj
- Noorul Islam Centre for Higher Education, Department of Computer Applications, Tamilnadu, India
| | - Victor Jose Marianthiran
- Vel Tech Multi Tech Dr. Rangarajan. Sakunthala Engineering College, Department of Artificial Intelligence and Data Science, Tamilnadu, India
| |
Collapse
|
13
|
Fishburn AT, Florio CJ, Klaessens TN, Prince B, Adia NAB, Lopez NJ, Beesabathuni NS, Becker SS, Cherkashchenko L, Haggard Arcé ST, Hoang V, Shiu TN, Richardson RB, Evans MJ, Rückert C, Shah PS. Microcephaly protein ANKLE2 promotes Zika virus replication. mBio 2025; 16:e0268324. [PMID: 39804047 PMCID: PMC11796389 DOI: 10.1128/mbio.02683-24] [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: 09/11/2024] [Accepted: 11/26/2024] [Indexed: 02/06/2025] Open
Abstract
Orthoflaviviruses are positive-sense single-stranded RNA viruses that hijack host proteins to promote their own replication. Zika virus (ZIKV) is infamous among orthoflaviviruses for its association with severe congenital birth defects, notably microcephaly. We previously mapped ZIKV-host protein interactions and identified the interaction between ZIKV non-structural protein 4A (NS4A) and host microcephaly protein ankyrin repeat and LEM domain-containing 2 (ANKLE2). Using a fruit fly model, we showed that NS4A induced microcephaly in an ANKLE2-dependent manner. Here, we explore the role of ANKLE2 in ZIKV replication to understand the biological significance of the interaction from a viral perspective. We observe that ANKLE2 localization is drastically shifted to sites of NS4A accumulation during infection and that knockout of ANKLE2 reduces ZIKV replication in multiple human cell lines. This decrease in virus replication is coupled with a moderate increase in innate immune activation. Using microscopy, we observe dysregulated formation of virus-induced endoplasmic reticulum rearrangements in ANKLE2 knockout cells. Knockdown of the ANKLE2 ortholog in Aedes aegypti cells also decreases virus replication, suggesting ANKLE2 is a beneficial replication factor across hosts. Finally, we show that NS4A from four other orthoflaviviruses physically interacts with ANKLE2 and is also beneficial to their replication. Thus, ANKLE2 likely promotes orthoflavivirus replication by regulating membrane rearrangements that serve to accelerate viral genome replication and protect viral dsRNA from immune detection. Taken together with our previous results, our findings indicate that ZIKV and other orthoflaviviruses hijack ANKLE2 for a conserved role in replication, and this drives unique pathogenesis for ZIKV since ANKLE2 has essential roles in developing tissues.IMPORTANCEZIKV is a major concern due to its association with birth defects, including microcephaly. We previously identified a physical interaction between ZIKV NS4A and host microcephaly protein ANKLE2. Mutations in ANKLE2 cause congenital microcephaly, and NS4A induces microcephaly in an ANKLE2-dependent manner. Here, we establish the role of ANKLE2 in ZIKV replication. Depletion of ANKLE2 from cells significantly reduces ZIKV replication and disrupts virus-induced membrane rearrangements. ANKLE2's ability to promote ZIKV replication is conserved in mosquito cells and for other related mosquito-borne orthoflaviviruses. Our data point to an overall model in which ANKLE2 regulates virus-induced membrane rearrangements to accelerate orthoflavivirus replication and avoid immune detection. However, ANKLE2's unique role in ZIKV NS4A-induced microcephaly is a consequence of ZIKV infection of important developing tissues in which ANKLE2 has essential roles.
Collapse
Affiliation(s)
- Adam T. Fishburn
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Cole J. Florio
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Thomas N. Klaessens
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Brian Prince
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada, USA
| | - Neil A. B. Adia
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Nicholas J. Lopez
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | | | - Sydney S. Becker
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Liubov Cherkashchenko
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Sophia T. Haggard Arcé
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Vivian Hoang
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - Traci N. Shiu
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
| | - R. Blake Richardson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Matthew J. Evans
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, University of Nevada, Reno, Nevada, USA
| | - Priya S. Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, California, USA
- Department of Chemical Engineering, University of California, Davis, California, USA
| |
Collapse
|
14
|
Hewitt CR, Wixon NJ, Gallegos A, Zhou Y, Huber VC, Killian MS. Inactivation of Zika Virus with Hydroxypropyl-Beta-Cyclodextrin. Vaccines (Basel) 2025; 13:79. [PMID: 39852858 PMCID: PMC11769224 DOI: 10.3390/vaccines13010079] [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: 11/01/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: Zika virus (ZIKV) infection is associated with life-threatening diseases in humans. To date, there are no available FDA-approved therapies or vaccines for the specific treatment or prevention of ZIKV infection. Variation in the ZIKV envelope protein (Env), along with its complex quaternary structure, presents challenges to synthetic approaches for developing an effective vaccine and broadly neutralizing antibodies (bnAbs). We hypothesized that beta-cyclodextrin (BCD) could be used to uniquely inactivate infectious ZIKV without disruption of Env. Methods: ZIKV was propagated in Vero cells and admixed with BCD. The BCD-treated ZIKV was evaluated for infectivity using immunofluorescence and quantitative RT-PCR (qRT-PCR) assays, for immunoreactivity in Western blots, structural integrity by electron microscopy, and immunogenicity in mice. Results: Here, we show that 200 mM BCD-treated ZIKV is non-infectious in cell culture, remains immunoreactive with an Env-specific antibody, retains its virion shape and size, and elicits the production of immunogen-specific antibodies in immunized mice. Conclusions: These results indicate that BCD can be used to safely inactivate ZIKV, and they provide insights for vaccine and antibody development.
Collapse
Affiliation(s)
- Cory R. Hewitt
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (C.R.H.); (V.C.H.)
| | - Nicholas J. Wixon
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (C.R.H.); (V.C.H.)
| | - Arthur Gallegos
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (C.R.H.); (V.C.H.)
| | - You Zhou
- Microscopy Core Research Facility, Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Victor C. Huber
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (C.R.H.); (V.C.H.)
| | - M. Scott Killian
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA; (C.R.H.); (V.C.H.)
- Department of Public Health, School of Health Sciences, University of South Dakota, Vermillion, SD 57069, USA
| |
Collapse
|
15
|
Song BH, Frank JC, Yun SI, Julander JG, Mason JB, Polejaeva IA, Davies CJ, White KL, Dai X, Lee YM. Comparison of Three Chimeric Zika Vaccine Prototypes Developed on the Genetic Background of the Clinically Proven Live-Attenuated Japanese Encephalitis Vaccine SA 14-14-2. Int J Mol Sci 2024; 26:195. [PMID: 39796052 PMCID: PMC11720029 DOI: 10.3390/ijms26010195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Zika virus (ZIKV) is a medically important mosquito-borne orthoflavivirus, but no vaccines are currently available to prevent ZIKV-associated disease. In this study, we compared three recombinant chimeric viruses developed as candidate vaccine prototypes (rJEV/ZIKVMR-766, rJEV/ZIKVP6-740, and rJEV/ZIKVPRVABC-59), in which the two neutralizing antibody-inducing prM and E genes from each of three genetically distinct ZIKV strains were used to replace the corresponding genes of the clinically proven live-attenuated Japanese encephalitis virus vaccine SA14-14-2 (rJEV). In WHO-certified Vero cells (a cell line suitable for vaccine production), rJEV/ZIKVP6-740 exhibited the slowest viral growth, formed the smallest plaques, and displayed a unique protein expression profile with the highest ratio of prM to cleaved M when compared to the other two chimeric viruses, rJEV/ZIKVMR-766 and rJEV/ZIKVPRVABC-59, as well as their vector, rJEV. In IFNAR-/- mice, an animal model of ZIKV infection, subcutaneous inoculation of rJEV/ZIKVP6-740 caused a low-level localized infection limited to the spleen, with no clinical signs of infection, weight loss, or mortality; in contrast, the other two chimeric viruses and their vector caused high-level systemic infections involving multiple organs, consistently leading to clear clinical signs of infection, rapid weight loss, and 100% mortality. Subsequently, subcutaneous immunization with rJEV/ZIKVP6-740 proved highly effective, offering complete protection against a lethal intramuscular ZIKV challenge 28 days after a single-dose immunization. This protection was specific to ZIKV prM/E and likely mediated by neutralizing antibodies targeting ZIKV prM/E. Therefore, our data indicate that the chimeric virus rJEV/ZIKVP6-740 is a highly promising vaccine prototype for developing a safe and effective vaccine for inducing neutralizing antibody-mediated protective immunity against ZIKV.
Collapse
Affiliation(s)
- Byung-Hak Song
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Jordan C. Frank
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Sang-Im Yun
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Justin G. Julander
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Jeffrey B. Mason
- Department of Veterinary Clinical and Life Sciences, College of Veterinary Medicine, Center for Integrated BioSystems, Utah State University, Logan, UT 84322, USA;
| | - Irina A. Polejaeva
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Christopher J. Davies
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Kenneth L. White
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Xin Dai
- Utah Agricultural Experiment Station, Utah State University, Logan, UT 84322, USA;
| | - Young-Min Lee
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| |
Collapse
|
16
|
Xu Y, Du J, Zhang K, Li J, Zou F, Li X, Meng Y, Chen Y, Tao L, Zhao F, Ma L, Shen B, Zhou D, Sun Y, Yan G, Zhu C. The Dual Resistance Mechanism of CYP325G4 and CYP6AA9 in Culex pipiens pallens Legs According to Transcriptome and Proteome Analysis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27150-27162. [PMID: 39604078 DOI: 10.1021/acs.jafc.4c05708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Mosquitoes within the Culex pipiens complex play a crucial role in human disease transmission. Insecticides, especially pyrethroids, are used to control these vectors. Mosquito legs are the main entry point and barrier for insecticides to gain their neuronal targets. However, the resistance mechanism in mosquito legs is unclear. Herein, we employed transcriptomic analyses and isobaric tags for relative and absolute quantitation techniques to investigate the resistance mechanism, focusing on Cx. pipiens legs. We discovered 2346 differentially expressed genes (DEGs) between deltamethrin-resistant (DR) and deltamethrin-sensitive (DS) mosquito legs, including 41 cytochrome P450 genes. In the same comparison, we identified 228 differentially expressed proteins (DEPs), including six cytochrome P450 proteins. Combined transcriptome and proteome analysis revealed only two upregulated P450 genes, CYP325G4 and CYP6AA9. The main clusters of DEGs and DEPs were associated with metabolic processes, such as cytochrome P450-mediated metabolism of drugs and xenobiotics. Transcription analysis revealed high CYP325G4 and CYP6AA9 expression in the DR strain at 72 h posteclosion compared with that in the DS strain, particularly in the legs. Mosquitoes knocked down for CYP325G4 were more sensitive to deltamethrin than the controls. CYP325G4 knockdown reduced the expression of several chlorinated hydrocarbon (CHC)-related genes, which altered the cuticle thickness and structure. Conversely, CYP6AA9 knockdown increased CHC gene expression without altering cuticle thickness and structure. P450 activity analysis demonstrated that CYP325G4 and CYP6AA9 contributed to metabolic resistance in the midgut and legs. This study identified CYP325G4 as a novel mosquito deltamethrin resistance factor, being involved in both metabolic and cuticular resistance mechanisms. The previously identified CYP6AA9 was investigated for its involvement in metabolic resistance and potential cuticular resistance in mosquito legs. These findings enhance our comprehension of resistance mechanisms, identifying P450s as promising targets for the future management of mosquito vector resistance, and laying a theoretical groundwork for mosquito resistance management.
Collapse
Affiliation(s)
- Yang Xu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiajia Du
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Kewei Zhang
- Department of Population Health & Disease Prevention, Joe C. Wen School of Population & Public Health, University of California, Irvine, California 92697, United States
| | - Jinze Li
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Feifei Zou
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xixi Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yufen Meng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Tao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fengming Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Ma
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Bo Shen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Dan Zhou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Yan Sun
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Guiyun Yan
- Department of Population Health & Disease Prevention, Joe C. Wen School of Population & Public Health, University of California, Irvine, California 92697, United States
| | - Changliang Zhu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
17
|
Pang WH, Wang BB, Huang YX, Zhou XM, Wu XW, Zeb MA, Zhang RH, Huang N, Li XL, Zheng CB, Xiao WL. Six new prenylated flavonoids from Dodonaea viscosa with anti-Zika virus activity. Fitoterapia 2024; 179:106264. [PMID: 39426434 DOI: 10.1016/j.fitote.2024.106264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Six new prenylated flavonoids, named visconaeas A-F (1-6), and eleven known isopentenyl flavonoids (7-17) were isolated from Dodonaea viscosa (L.) Jacq. The structures of the separated compounds were determined through comprehensive spectral analysis and quantum chemical calculations. These compounds were tested for their anti-Zika virus and cytotoxicity activities. The results indicated that compound 4 showed low cytotoxicity and strong anti-Zika virus potential with EC50 16.34 μM.
Collapse
Affiliation(s)
- Wen-Hui Pang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Bin-Bao Wang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, People's Republic of China; Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, People's Republic of China
| | - Yong-Xiang Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Xue-Mei Zhou
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Xue-Wen Wu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Muhammad Aurang Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Rui-Han Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Ning Huang
- Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, People's Republic of China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China.
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, People's Republic of China; Yunnan Vaccine Laboratory, Kunming 650500, People's Republic of China; College of Modern Biomedical Industry, Kunming Medical University, Kunming, Yunnan 650500, People's Republic of China.
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Key Laboratory of Research and Development for Natural Products, School of Chemical Science and Technology, School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China; Southwest United Graduate School, Kunming 650592, People's Republic of China.
| |
Collapse
|
18
|
Yang C, Chen W, Huang Y. Long non-coding RNA SUN2-AS1 acts as a negative regulator of ISGs transcription to promote flavivirus infection. Virology 2024; 600:110245. [PMID: 39288611 DOI: 10.1016/j.virol.2024.110245] [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: 07/02/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Recent studies highlight the critical involvement of long non-coding RNAs (lncRNAs) in modulating viral replication and immune responses, yet their specific roles in flavivirus infections remain underexplored. Our study has identified lncRNA SUN2-AS1, which is significantly upregulated in response to flavivirus infection in A549, Huh7 cells, and monocyte-differentiated macrophages (MDMs). SUN2-AS1 interacts with the transcription factors NF-κB and STAT1, andits expression is induced by ZIKV RNA via the type I interferon (IFN) pathway. Notably, SUN2-AS1 enhances the infection of flaviviruses, including ZIKV, DENV2, and JEV, while showing no effect on VSV or HSV-1 infections. Mechanistically, SUN2-AS1 exerts a proviral effect by inhibiting the transcription of interferon-stimulated genes (ISGs). These findings uncover a novel mechanism by which lncRNAs facilitate flavivirus propagation and highlight SUN2-AS1 as a potential target for antiviral therapeutic strategies.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; Guangxi Hospital Division of the First Affiliated Hospital, Sun Yat-sen University, Nanning, 530022, China
| | - Weikang Chen
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yanxia Huang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
19
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
20
|
Zhang L, Wang H, Han C, Dong Q, Yan J, Guo W, Shan C, Zhao W, Chen P, Huang R, Wu Y, Chen Y, Qin Y, Chen M. AMFR-mediated Flavivirus NS2A ubiquitination subverts ER-phagy to augment viral pathogenicity. Nat Commun 2024; 15:9578. [PMID: 39505910 PMCID: PMC11541587 DOI: 10.1038/s41467-024-54010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
Flaviviruses strategically utilize the endoplasmic reticulum (ER) in their replication cycles. However, the role of ER autophagy (ER-phagy) in viral replication process remains poorly understood. Here, we reveal that prolonged Zika virus (ZIKV) infection results from the degradation of ER-phagy receptor FAM134B, facilitated by viral NS2A protein. Mechanistically, ER-localized NS2A undergoes K48-linked polyubiquitination at lysine (K) 56 by E3 ligase AMFR. Ubiquitinated NS2A binds to FAM134B and AMFR orchestrates the degradation of NS2A-FAM134B complexes. AMFR-catalyzed NS2A ubiquitination not only targets FAM134B degradation but also hinders the FAM134B-AMFR axis. Notably, a recombinant ZIKV mutant (ZIKV-NS2AK56R), lacking ubiquitination and ER-phagy inhibition, exhibits attenuation in ZIKV-induced microcephalic phenotypes in human brain organoids and replicates less efficiently, resulting in weakened pathogenesis in mouse models. In this work, our mechanistic insights propose that flaviviruses manipulate ER-phagy to modulate ER turnover, driving viral infection. Furthermore, AMFR-mediated flavivirus NS2A ubiquitination emerges as a potential determinant of viral pathogenecity.
Collapse
Affiliation(s)
- Linliang Zhang
- School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Hongyun Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Chao Han
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Qi Dong
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Jie Yan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Weiwei Guo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430072, China
- University of the Chinese Academy of Sciences, Beijing, 100039, China
| | - Chao Shan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430072, China
- University of the Chinese Academy of Sciences, Beijing, 100039, China
| | - Wen Zhao
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, 430072, China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan, 430072, China
| | - Rui Huang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430072, China
| | - Ying Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430072, China
| | - Yu Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Mingzhou Chen
- School of Life Sciences, Hubei University, Wuhan, 430062, China.
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
21
|
Boytz R, Laurent-Rolle M. Balancing acts: The posttranslational modification tightrope of flavivirus replication. PLoS Pathog 2024; 20:e1012626. [PMID: 39466723 PMCID: PMC11516179 DOI: 10.1371/journal.ppat.1012626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
Posttranslational modifications (PTMs) such as phosphorylation, ubiquitination, SUMOylation, and ISGylation are involved in various cellular pathways, including innate immunity and disease processes. Many viruses have developed sophisticated mechanisms to modulate these host PTMs, either by inhibiting the interferon pathway or by enhancing the stability and function of viral proteins essential for replication. In this Pearl, we review the literature on how flaviviruses are impacted by and exploit posttranslational modifications to their advantage.
Collapse
Affiliation(s)
- RuthMabel Boytz
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Maudry Laurent-Rolle
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
22
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
23
|
Yan Y, Yang H, Yang Y, Wang J, Zhou Y, Tang C, Li B, Huang Q, An R, Liang X, Lin D, Yu W, Fan C, Lu S. The inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in immunocompromised AG129 mice. MOLECULAR BIOMEDICINE 2024; 5:30. [PMID: 39095588 PMCID: PMC11297010 DOI: 10.1186/s43556-024-00195-x] [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: 02/19/2024] [Accepted: 07/04/2024] [Indexed: 08/04/2024] Open
Abstract
Zika virus, a mosquito-borne arbovirus, has repeatedly caused large pandemics with symptoms worsening from mild and self-limiting diseases to Guillain-Barré syndrome in adults and fetal microcephaly in newborns. In recent years, Zika virus diseases have posed a serious threat to human health. The shortage of susceptible small animal models makes it difficult to study pathogenic mechanisms and evaluate potential therapies for Zika virus infection. Therefore, we chose immunocompromised mice (AG129 mice) deficient in IFN-α/β and IFN-γ receptors, which can abolish the innate immune system that prevents Zika virus infection early. AG129 mice were infected with the Zika virus, and this mouse model exhibited replication dynamics, tissue tropism, pathological lesion and immune activation of the Zika virus. Our results suggest that the inoculum dose of Zika virus can affect the viral replication dynamics, cytokine responses and survival rate in AG129 mice. By testing the potential antiviral drug favipiravir, several critical indicators, including replication dynamics and survival rates, were identified in AG129 mice after Zika virus infection. It is suggested that the model is reliable for drug evaluation. In brief, this model provides a potential platform for studies of the infectivity, virulence, and pathogenesis of the Zika virus. Moreover, the development of an accessible mouse model of Zika virus infection will expedite the research and deployment of therapeutics and vaccines.
Collapse
Affiliation(s)
- Yuhuan Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Hao Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yun Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Junbin Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yanan Zhou
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Cong Tang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Bai Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Qing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Ran An
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xiaoming Liang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Dongdong Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Wenhai Yu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
| | - Changfa Fan
- National Rodent Laboratory Animal Resources Center, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, 102629, China.
| | - Shuaiyao Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, China.
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Kunming, China.
| |
Collapse
|
24
|
Machmouchi D, Courageot MP, Ogire E, Redecke L, Kohl A, Desprès P, Roche M. The NS1 protein of contemporary West African Zika virus potentiates viral replication and reduces innate immune activation. PLoS Negl Trop Dis 2024; 18:e0012146. [PMID: 39178324 PMCID: PMC11376516 DOI: 10.1371/journal.pntd.0012146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/05/2024] [Accepted: 08/01/2024] [Indexed: 08/25/2024] Open
Abstract
Mosquito-borne Zika virus (ZIKV) from sub-Saharan Africa has recently gained attention due to its epidemic potential and its capacity to be highly teratogenic. To improve our knowledge on currently circulating strains of African ZIKV, we conducted protein sequence alignment and identified contemporary West Africa NS1 (NS1CWA) protein as a highly conserved viral protein. Comparison of NS1CWA with the NS1 of the historical African ZIKV strain MR766 (NS1MR766), revealed seven amino acid substitutions. The effects of NS1 mutations on protein expression, virus replication, and innate immune activation were assessed in human cells using recombinant NS1 proteins and a chimeric viral clone MR766 with NS1CWA replacing NS1MR766. Our data indicated higher secretion efficiency of NS1CWA compared to NS1MR766 associated with a change in subcellular distribution. A chimeric MR766 virus with NS1CWA instead of authentic protein displayed a greater viral replication efficiency, leading to more pronounced cell death compared to parental virus. Enhanced viral growth was associated with reduced activation of innate immunity. Our data raise questions of the importance of NS1 protein in the pathogenicity of contemporary ZIKV from sub-Saharan Africa and point to differences within viral strains of African lineage.
Collapse
Affiliation(s)
- Dana Machmouchi
- Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI, Sainte-Clotilde, La Réunion, France
| | | | - Eva Ogire
- Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI, Sainte-Clotilde, La Réunion, France
| | - Lars Redecke
- University of Luebeck, Institute of Biochemistry, Luebeck, Germany
- Deutsches Elektronen Synchrotron (DESY), Photon Science, Hamburg, Germany
| | - Alain Kohl
- Centre for Neglected Tropical Diseases, Departments of Tropical Disease Biology and Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United-Kingdom
| | - Philippe Desprès
- Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI, Sainte-Clotilde, La Réunion, France
| | - Marjolaine Roche
- Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI, Sainte-Clotilde, La Réunion, France
| |
Collapse
|
25
|
Gilbert-Jaramillo J, Komarasamy TV, Balasubramaniam VR, Heather LC, James WS. Targeting glucose metabolism with dichloroacetate (DCA) reduces zika virus replication in brain cortical progenitors at different stages of maturation. Antiviral Res 2024; 228:105933. [PMID: 38851593 DOI: 10.1016/j.antiviral.2024.105933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
The underlying threat of new Zika virus (ZIKV) outbreaks remains, as no vaccines or therapies have yet been developed. In vitro research has shown that glycolysis is a key factor to enable sustained ZIKV replication in neuroprogenitors. However, neither in vivo nor clinical investigation of glycolytic modulators as potential therapeutics for ZIKV-related fetal abnormalities has been conducted. Accordingly, we tested the therapeutic potential of metabolic modulators in relevant in vitro systems comprising two pools of neuroprogenitors (NPCs), which resemble early and late stages of pregnancy. Effective doses of metabolic modulators [3.0 μM] dimethyl fumarate (DMF), [3.2 mM] dichloroacetate (DCA), and [6.3 μM] VER-246608 were determined for these cells by their effect on lactate release, pyruvate dehydrogenase (PDH) activity and cell survival. The drugs were used in a 24h pre-treatment and kept throughout ZIKV infection of NPCs. Drug effects and ZIKV replication were assessed at 24- and 56-h post-infection. In early NPCs treated with DMF, DCA and VER-246608, there was a significant reduction in the extracellular release of ZIKV potentially by PDH-mediated increased mitochondrial oxidation of glucose. Out of the three drugs, only DCA was observed to reduce viral replication in late NPCs treated with DCA. Altogether, our findings suggest that reduction of anaerobic glycolysis could be of therapeutic potential against ZIKV-related fetal abnormalities and that clinical translation should consider the use of specific glycolytic modulators over different trimesters.
Collapse
Affiliation(s)
- Javier Gilbert-Jaramillo
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Vinod Rmt Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - William S James
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| |
Collapse
|
26
|
Liu T, Li M, Tian Y, Dong Y, Liu N, Wang Z, Zhang H, Zheng A, Cui C. Immunogenicity and safety of a self-assembling ZIKV nanoparticle vaccine in mice. Int J Pharm 2024; 660:124320. [PMID: 38866086 DOI: 10.1016/j.ijpharm.2024.124320] [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: 02/14/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that highly susceptibly causes Guillain-Barré syndrome and microcephaly in newborns. Vaccination is one of the most effective measures for preventing infectious diseases. However, there is currently no approved vaccine to prevent ZIKV infection. Here, we developed nanoparticle (NP) vaccines by covalently conjugating self-assembled 24-subunit ferritin to the envelope structural protein subunit of ZIKV to achieve antigen polyaggregation. The immunogenicityof the NP vaccine was evaluated in mice. Compared to monomer vaccines, the NP vaccine achieved effective antigen presentation, promoted the differentiation of follicular T helper cells in lymph nodes, and induced significantly greater antigen-specific humoral and cellular immune responses. Moreover, the NP vaccine enhanced high-affinity antigen-specific IgG antibody levels, increased secretion of the cytokines IL-4 and IFN-γ by splenocytes, significantly activated T/B lymphocytes, and improved the generation of memory T/B cells. In addition, no significant adverse reactions occurred when NP vaccine was combined with adjuvants. Overall, ferritin-based NP vaccines are safe and effective ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Meng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yuhan Dong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Nan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China.
| |
Collapse
|
27
|
Alharbi M, Alshammari A, Alsabhan JF, Alzarea SI, Alshammari T, Alasmari F, Alasmari AF. A novel vaccine construct against Zika virus fever: insights from epitope-based vaccine discovery through molecular modeling and immunoinformatics approaches. Front Immunol 2024; 15:1426496. [PMID: 39050858 PMCID: PMC11267680 DOI: 10.3389/fimmu.2024.1426496] [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: 05/02/2024] [Accepted: 06/05/2024] [Indexed: 07/27/2024] Open
Abstract
The Zika virus (ZIKV) is an emerging virus associated with the Flaviviridae family that mainly causes infection in pregnant women and leads to several abnormalities during pregnancy. This virus has unique properties that may lead to pathological diseases. As the virus has the ability to evade immune response, a crucial effort is required to deal with ZIKV. Vaccines are a safe means to control different pathogenic infectious diseases. In the current research, a multi-epitope-based vaccination against ZIKV is being designed using in silico methods. For the epitope prediction and prioritization phase, ZIKV polyprotein (YP_002790881.1) and flavivirus polyprotein (>YP_009428568.1) were targeted. The predicted B-cell epitopes were used for MHC-I and MHC-II epitope prediction. Afterward, several immunoinformatics filters were applied and nine (REDLWCGSL, MQDLWLLRR, YKKSGITEV, TYTDRRWCF, RDAFPDSNS, KPSLGLINR, ELIGRARVS, AITQGKREE, and EARRSRRAV) epitopes were found to be probably antigenic in nature, non-allergenic, non-toxic, and water soluble without any toxins. Selected epitopes were joined using a particular GPGPG linker to create the base vaccination for epitopes, and an extra EAAAK linker was used to link the adjuvant. A total of 312 amino acids with a molecular weight (MW) of 31.62762 and an instability value of 34.06 were computed in the physicochemical characteristic analysis, indicating that the vaccine design is stable. The molecular docking analysis predicted a binding energy of -329.46 (kcal/mol) for TLR-3 and -358.54 (kcal/mol) for TLR-2. Moreover, the molecular dynamics simulation analysis predicted that the vaccine and receptor molecules have stable binding interactions in a dynamic environment. The C-immune simulation analysis predicted that the vaccine has the ability to generate both humoral and cellular immune responses. Based on the design, the vaccine construct has the best efficacy to evoke immune response in theory, but experimental analysis is required to validate the in silico base approach and ensure its safety.
Collapse
Affiliation(s)
- Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Jawza F. Alsabhan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, Saudi Arabia
| | - Talal Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Dionne E, Machiavello Roman F, Farhadian S. Climate Change and Meningoencephalitis in the Americas: A Brewing Storm. Curr Infect Dis Rep 2024; 26:189-196. [DOI: 10.1007/s11908-024-00843-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 01/04/2025]
|
29
|
Aguiar GRF, da Silva GB, Ramalho JDAM, Srisawat N, Daher EDF. Common arboviruses and the kidney: a review. J Bras Nefrol 2024; 46:e20230168. [PMID: 39074252 PMCID: PMC11287847 DOI: 10.1590/2175-8239-jbn-2023-0168en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/17/2024] [Indexed: 07/31/2024] Open
Abstract
Arboviruses are endemic in several countries and represent a worrying public health problem. The most important of these diseases is dengue fever, whose numbers continue to rise and have reached millions of annual cases in Brazil since the last decade. Other arboviruses of public health concern are chikungunya and Zika, both of which have caused recent epidemics, and yellow fever, which has also caused epidemic outbreaks in our country. Like most infectious diseases, arboviruses have the potential to affect the kidneys through several mechanisms. These include the direct action of the viruses, systemic inflammation, hemorrhagic phenomena and other complications, in addition to the toxicity of the drugs used in treatment. In this review article, the epidemiological aspects of the main arboviruses in Brazil and other countries where these diseases are endemic, clinical aspects and the main laboratory changes found, including changes in renal function, are addressed. It also describes how arboviruses behave in kidney transplant patients. The pathophysiological mechanisms of kidney injury associated with arboviruses are described and finally the recommended treatment for each disease and recommendations for kidney support in this context are given.
Collapse
Affiliation(s)
- Gabriel Rotsen Fortes Aguiar
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
| | - Geraldo Bezerra da Silva
- Universidade de Fortaleza, Centro de Ciências da Saúde, Curso de Medicina, Fortaleza, CE, Brazil
| | - Janaína de Almeida Mota Ramalho
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
- Universidade de Fortaleza, Centro de Ciências da Saúde, Curso de Medicina, Fortaleza, CE, Brazil
| | - Nattachai Srisawat
- Chulalongkorn University, Faculty of Medicine, Department of Medicine, Division of Nephrology, Center of Excellence for Critical Care Nephrology, and Tropical Medicine Cluster, Bangkok, Tailândia
| | - Elizabeth de Francesco Daher
- Universidade Federal do Ceará, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Departamento de Medicina Interna, Fortaleza, CE, Brazil
| |
Collapse
|
30
|
Huang GG, Wang HY, Wang XH, Yang T, Zhang XM, Feng CL, Zhao WM, Tang W. Atranorin inhibits Zika virus infection in human glioblastoma cell line SNB-19 via targeting Zika virus envelope protein. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155343. [PMID: 38290230 DOI: 10.1016/j.phymed.2024.155343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/26/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Zika virus (ZIKV) is a single-stranded RNA flavivirus transmitted by mosquitoes. Its infection is associated with neurological complications such as neonatal microcephaly and adult Guillain-Barré syndrome, posing a serious threat to the health of people worldwide. Therefore, there is an urgent need to develop effective anti-ZIKV drugs. Atranorin is a lichen secondary metabolite with a wide range of biological activities, including anti-inflammatory, antibacterial and antioxidant, etc. However, the antiviral activity of atranorin and underlying mechanism has not been fully elucidated. PURPOSE We aimed to determine the anti-ZIKV activity of atranorin in human glioma cell line SNB-19 and investigate the potential mechanism from the perspective of viral life cycle and the host cell functions. METHODS We first established ZIKV-infected human glioma cells (SNB-19) model and used Western Blot, RT-qPCR, immunofluorescence, fluorescence-activated cell sorting (FACS) and plaque assay to evaluate the anti-ZIKV activity of atranorin. Then we assessed the regulation effect of atranorin on ZIKV induced IFN signal pathway activation by RT-qPCR. Afterward, we introduced time-of-addition assay, viral adsorption assay, viral internalization assay and transferrin uptake assay to define which step of ZIKV lifecycle is influenced by atranorin. Finally, we performed virus infectivity assay, molecular docking and thermal shift assay to uncover the target protein of atranorin on ZIKV. RESULTS Our study showed that atranorin could protect SNB-19 cells from ZIKV infection, as evidenced by inhibited viral protein expression and progeny virus yield. Meanwhile, atranorin attenuated the activation of IFN signal pathway and downstream inflammatory response that induced by ZIKV infection. The results of time-of-addition assay indicated that atranorin acted primarily by disturbing the viral entry process. After ruling out the effect of atranorin on AXL receptor tyrosine kinase (AXL) dependent virus adsorption and clathrin-mediated endocytosis, we confirmed that atranorin directly targeted the viral envelope protein and lowered ZIKV infectivity by thermal shift assay and virus infectivity assay respectively. CONCLUSION We found atranorin inhibits ZIKV infection in SNB-19 cells via targeting ZIKV envelope protein. Our study provided an experimental basis for the further development of atranorin and a reference for antiviral drug discovery from natural resources.
Collapse
Affiliation(s)
- Guan-Gen Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Hao-Yu Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiao-Han Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tao Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiao-Meng Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Chun-Lan Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Wei-Min Zhao
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Wei Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
31
|
Formanski JP, Ngo HD, Grunwald V, Pöhlking C, Jonas JS, Wohlers D, Schwalbe B, Schreiber M. Transduction Efficiency of Zika Virus E Protein Pseudotyped HIV-1 gfp and Its Oncolytic Activity Tested in Primary Glioblastoma Cell Cultures. Cancers (Basel) 2024; 16:814. [PMID: 38398205 PMCID: PMC10887055 DOI: 10.3390/cancers16040814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
The development of new tools against glioblastoma multiforme (GBM), the most aggressive and common cancer originating in the brain, remains of utmost importance. Lentiviral vectors (LVs) are among the tools of future concepts, and pseudotyping offers the possibility of tailoring LVs to efficiently transduce and inactivate GBM tumor cells. Zika virus (ZIKV) has a specificity for GBM cells, leaving healthy brain cells unharmed, which makes it a prime candidate for the development of LVs with a ZIKV coat. Here, primary GBM cell cultures were transduced with different LVs encased with ZIKV envelope variants. LVs were generated by using the pNLgfpAM plasmid, which produces the lentiviral, HIV-1-based, core particle with GFP (green fluorescent protein) as a reporter (HIVgfp). Using five different GBM primary cell cultures and three laboratory-adapted GBM cell lines, we showed that ZIKV/HIVgfp achieved a 4-6 times higher transduction efficiency compared to the commonly used VSV/HIVgfp. Transduced GBM cell cultures were monitored over a period of 9 days to identify GFP+ cells to study the oncolytic effect due to ZIKV/HIVgfp entry. Tests of GBM tumor specificity by transduction of GBM tumor and normal brain cells showed a high specificity for GBM cells.
Collapse
Affiliation(s)
- Jan Patrick Formanski
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Hai Dang Ngo
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Vivien Grunwald
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Celine Pöhlking
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Jana Sue Jonas
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Dominik Wohlers
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| | - Birco Schwalbe
- Department of Neurosurgery, Asklepios Klinik Nord, Standort Heidberg, 22417 Hamburg, Germany;
| | - Michael Schreiber
- Department of Virology, LG Schreiber, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany (H.D.N.); (V.G.); (C.P.); (J.S.J.); (D.W.)
| |
Collapse
|
32
|
Mirza AH, Bram Y, Schwartz RE, Jaffrey SR. SCARPET: site-specific quantification of methylated and nonmethylated adenosines reveals m 6A stoichiometry. RNA (NEW YORK, N.Y.) 2024; 30:308-324. [PMID: 38190635 PMCID: PMC10870371 DOI: 10.1261/rna.079776.123] [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: 07/18/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
m6A has different stoichiometry at different positions in different mRNAs. However, the exact stoichiometry of m6A is difficult to measure. Here, we describe SCARPET (site-specific cleavage and radioactive-labeling followed by purification, exonuclease digestion, and thin-layer chromatography), a simple and streamlined biochemical assay for quantifying m6A at any specific site in any mRNA. SCARPET involves a site-specific cleavage of mRNA immediately 5' of an adenosine site in an mRNA. This site is radiolabeled with 32P, and after a series of steps to purify the RNA and to remove nonspecific signals, the nucleotide is resolved by TLC to visualize A and m6A at this site. Quantification of these spots reveals the m6A stoichiometry at the site of interest. SCARPET can be applied to poly(A)-enriched RNA, or preferably purified mRNA, which produces more accurate m6A stoichiometry measurements. We show that sample processing steps of SCARPET can be performed in a single day, and results in a specific and accurate measurement of m6A stoichiometry at specific sites in mRNA. Using SCARPET, we measure exact m6A stoichiometries in specific mRNAs and show that Zika genomic RNA lacks m6A at previously mapped sites. SCARPET will be useful for testing specific sites for their m6A stoichiometry and to assess how m6A stoichiometry changes in different conditions and cellular contexts.
Collapse
Affiliation(s)
- Aashiq H Mirza
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Yaron Bram
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
- Department of Physiology Biophysics and Systems Biology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Samie R Jaffrey
- Department of Pharmacology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| |
Collapse
|
33
|
Peng ZY, Yang S, Lu HZ, Wang LM, Li N, Zhang HT, Xing SY, Du YN, Deng SQ. A review on Zika vaccine development. Pathog Dis 2024; 82:ftad036. [PMID: 38192053 PMCID: PMC10901608 DOI: 10.1093/femspd/ftad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Zika virus (ZIKV), which belongs to the Flavivirus family, is mainly transmitted via the bite of Aedes mosquitoes. In newborns, ZIKV infection can cause severe symptoms such as microcephaly, while in adults, it can lead to Guillain‒Barré syndrome (GBS). Due to the lack of specific therapeutic methods against ZIKV, the development of a safe and effective vaccine is extremely important. Several potential ZIKV vaccines, such as live attenuated, inactivated, nucleic acid, viral vector, and recombinant subunit vaccines, have demonstrated promising outcomes in clinical trials involving human participants. Therefore, in this review, the recent developmental progress, advantages and disadvantages of these five vaccine types are examined, and practical recommendations for future development are provided.
Collapse
Affiliation(s)
- Zhe-Yu Peng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Song Yang
- Institute of Agro-products Processing, Anhui Academy of Agricultural Sciences, Hefei 230031, Anhui, China
| | - Hong-Zheng Lu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lin-Min Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ni Li
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hai-Ting Zhang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Si-Yu Xing
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yi-Nan Du
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Sheng-Qun Deng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
34
|
Dhanushkumar T, Selvam PK, M E S, Vasudevan K, C GPD, Zayed H, Kamaraj B. Rational design of a multivalent vaccine targeting arthropod-borne viruses using reverse vaccinology strategies. Int J Biol Macromol 2024; 258:128753. [PMID: 38104690 DOI: 10.1016/j.ijbiomac.2023.128753] [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: 07/29/2023] [Revised: 11/17/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
Viruses transmitted by arthropods, such as Dengue, Zika, and Chikungunya, represent substantial worldwide health threats, particularly in countries like India. The lack of approved vaccines and effective antiviral therapies calls for developing innovative strategies to tackle these arboviruses. In this study, we employed immunoinformatics methodologies, incorporating reverse vaccinology, to design a multivalent vaccine targeting the predominant arboviruses. Epitopes of B and T cells were recognized within the non-structural proteins of Dengue, Zika, and Chikungunya viruses. The predicted epitopes were enhanced with adjuvants β-defensin and RS-09 to boost the vaccine's immunogenicity. Sixteen distinct vaccine candidates were constructed, each incorporating epitopes from all three viruses. FUVAC-11 emerged as the most promising vaccine candidate through molecular docking and molecular dynamics simulations, demonstrating favorable binding interactions and stability. Its effectiveness was further evaluated using computational immunological studies confirming strong immune responses. The in silico cloning performed using the pET-28a(+) plasmid facilitates the future experimental implementation of this vaccine candidate, paving the way for potential advancements in combating these significant arboviral threats. However, further in vitro and in vivo studies are warranted to confirm the results obtained in this computational study, which highlights the effectiveness of immunoinformatics and reverse vaccinology in creating vaccines against major Arboviruses, offering a promising model for developing vaccines for other vector-borne diseases and enhancing global health security.
Collapse
Affiliation(s)
- T Dhanushkumar
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Prasanna Kumar Selvam
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Santhosh M E
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India.
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India.
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Balu Kamaraj
- Department of Dental Education, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
35
|
de Sales-Neto JM, Madruga Carvalho DC, Arruda Magalhães DW, Araujo Medeiros AB, Soares MM, Rodrigues-Mascarenhas S. Zika virus: Antiviral immune response, inflammation, and cardiotonic steroids as antiviral agents. Int Immunopharmacol 2024; 127:111368. [PMID: 38103408 DOI: 10.1016/j.intimp.2023.111368] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus first reported from humans in Nigeria in 1954. The first outbreak occurred in Micronesia followed by an outbreak in French Polynesia and another in Brazil when the virus was associated with numerous cases of severe neurological manifestations such as Guillain-Barre syndrome in adults and congenital zika syndrome in fetuses, particularly congenital microcephaly. Innate immunity is the first line of defense against ZIKV through triggering an antiviral immune response. Along with innate immune responses, a sufficient balance between anti- and pro-inflammatory cytokines and the amount of these cytokines are triggered to enhance the antiviral responses. Here, we reviewed the complex interplay between the mediators and signal pathways that coordinate antiviral immune response and inflammation as a key to understanding the development of the underlying diseases triggered by ZIKV. In addition, we summarize current and new therapeutic strategies for ZIKV infection, highlighting cardiotonic steroids as antiviral drugs for the development of this agent.
Collapse
Affiliation(s)
- José Marreiro de Sales-Neto
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | | | - Mariana Mendonça Soares
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil.
| |
Collapse
|
36
|
Calvet GA, Kara EO, Bôtto-Menezes CHA, da Costa Castilho M, de Oliveira Franca RF, Habib N, Neto AM, Pereira GFM, Giozza SP, Bermúdez XPD, Fernandes TJ, Modjarrad K, Brasil P, Broutet NJN, de Filippis AMB. Detection and persistence of Zika virus in body fluids and associated factors: a prospective cohort study. Sci Rep 2023; 13:21557. [PMID: 38057382 PMCID: PMC10700488 DOI: 10.1038/s41598-023-48493-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
This study aimed to analyze the detection and duration of the Zika virus (ZIKV) in plasma, urine, saliva, sweat, rectal swabs, vaginal secretions, breast milk, and semen and to explore risk factors associated with prolonged viral persistence. A prospective cohort study of symptomatic patients and their household contacts was conducted in Brazil from July 2017 to June 2019. A total of 260 individuals (184 women and 76 men) with confirmed ZIKV infection were enrolled and followed up for 12 months. ZIKV RNA was present in all body fluid specimens and detectable for extended periods in urine, sweat, rectal swabs, and semen. The longest detection duration was found in semen, with high viral loads in the specimens. ZIKV RNA clearance was associated with several factors, including age, sex, education level, body mass index, non-purulent conjunctivitis, joint pain, and whether the participant had a history of yellow fever vaccination. The influence of each of these factors on the low or fast viral clearance varied according to the specific body fluid under investigation. Recurrent ZIKV detection events after total viral clearance were observed in the cohort. Our findings provide valuable insights into the persistence and potential recurrence of ZIKV infection, highlighting the need for continued monitoring and follow-up of individuals infected with ZIKV and for effective prevention measures to reduce the risk of transmission.
Collapse
Affiliation(s)
- Guilherme Amaral Calvet
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Edna Oliveira Kara
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Camila Helena Aguiar Bôtto-Menezes
- Department of Malaria, Tropical Medicine Foundation Doctor Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
- School of Health Sciences, Amazonas State University (UEA), Manaus, Amazonas, Brazil
| | - Marcia da Costa Castilho
- Department of Malaria, Tropical Medicine Foundation Doctor Heitor Vieira Dourado (FMT-HVD), Manaus, Amazonas, Brazil
| | | | - Ndema Habib
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Armando Menezes Neto
- Department of Virology and Experimental Therapy, Institute Aggeu Magalhães, Oswaldo Cruz Foundation, Recife, Pernambuco, Brazil
| | - Gerson Fernando Mendes Pereira
- Department of HIV/AIDS, Tuberculosis, Viral Hepatitis and Sexually Transmitted Infections (DATHI), Ministry of Health, Brasília, Brazil
| | - Silvana Pereira Giozza
- Department of HIV/AIDS, Tuberculosis, Viral Hepatitis and Sexually Transmitted Infections (DATHI), Ministry of Health, Brasília, Brazil
| | | | - Tatiana Jorge Fernandes
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Patrícia Brasil
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Maria Bispo de Filippis
- Flavivirus Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
37
|
Tu WC, Huang YX, Li B, Jiang YJ, Yang QY, Zeb MA, Yang PY, Wang HJ, Li XL, Xiao WL, Zheng CB, Liu MF. Wulfenioidins D-N, Structurally Diverse Diterpenoids with Anti-Zika Virus Activity Isolated from Orthosiphon wulfenioides. JOURNAL OF NATURAL PRODUCTS 2023; 86:2348-2359. [PMID: 37737089 DOI: 10.1021/acs.jnatprod.3c00543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Eleven diterpenoids, wulfenioidins D-N (1-11), classified into five distinct carbon skeletons with one unreported framework, and four modified abietane diterpenoids were isolated from the whole plant of Orthosiphon wulfenioides. The structures and absolute configurations were characterized by spectroscopic methods, single-crystal X-ray diffraction, and electronic circular dichroism analyses. Compounds 3 and 5 exhibited activity against Zika virus (ZIKV) with EC50 values of 8.07 and 8.50 μM, respectively, and showed no significant cytotoxicity toward Vero cells at 100 μM. Western blot and immunofluorescence experiments showed that compounds 3 and 5 interfered with the replication of the ZIKV by inhibiting the expression of the ZIKV envelope (E) protein.
Collapse
Affiliation(s)
- Wen-Chao Tu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Yong-Xiang Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Yunnan Vaccine Laboratory, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Bo Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Ying-Jie Jiang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Yunnan Vaccine Laboratory, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Quan-Yu Yang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Muhammad Aurang Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Peng-Yun Yang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Hui-Juan Wang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Provincial Center for Research & Development of Natural Products, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Pharmacy and School of Chemical Science and Technology, Yunnan University, Kunming, 650091, People's Republic of China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Yunnan Vaccine Laboratory, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Mei-Feng Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| |
Collapse
|
38
|
Liu Y, Jiang L, Sun X, Song Y, Liu Y, Zhang L. Interplay between TRIM7 and antiviral immunity. Front Cell Infect Microbiol 2023; 13:1256882. [PMID: 37719674 PMCID: PMC10500128 DOI: 10.3389/fcimb.2023.1256882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
TRIM7 has been demonstrated to have significant roles in promoting host defense against viral infections and regulating immune signaling pathways. As an E3 ubiquitin ligase, it catalyzes the ubiquitination of various substrates, including adaptor proteins (MAVS and STING) and transcription factors (NF-κB and IRF3), thereby exerting positive or negative regulation on immune signal pathways. However, viruses have developed immune evasion mechanisms to counteract TRIM7. Some viruses can inhibit TRIM7 function by targeting it for degradation or sequestering it away from its targets. Moreover, TRIM7 may even facilitate viral infection by ubiquitinating viral proteins, including envelope proteins that are critical for tissue and species tropism. A comprehensive understanding of the interaction between TRIM7 and antiviral immunity is crucial for the development of innovative treatments for viral diseases.
Collapse
Affiliation(s)
- Yiyang Liu
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lu Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuemeng Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yixuan Song
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yihan Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
39
|
Barbosa MD, Costa A, Prieto-Oliveira P, Andreata-Santos R, Peter CM, Zanotto PMA, Janini LMR. Proposal of Model for Evaluation of Viral Kinetics of African/Asian/Brazilian- Zika virus Strains (Step Growth Curve) in Trophoblastic Cell Lines. Viruses 2023; 15:1446. [PMID: 37515134 PMCID: PMC10386092 DOI: 10.3390/v15071446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
The Zika virus (ZIKV) epidemic brought new discoveries regarding arboviruses, especially flaviviruses, as ZIKV was described as sexually and vertically transmitted. The latter shows severe consequences for the embryo/fetus, such as congenital microcephaly and deficiency of the neural system, currently known as Congenital ZIKV Syndrome (CZS). To better understand ZIKV dynamics in trophoblastic cells present in the first trimester of pregnancy (BeWo, HTR-8, and control cell HuH-7), an experiment of viral kinetics was performed for African MR766 low passage and Asian-Brazilian IEC ZIKV lineages. The results were described independently and demonstrated that the three placental cells lines are permissive and susceptible to ZIKV. We noticed cytopathic effects that are typical in in vitro viral infection in BeWo and HTR-8. Regarding kinetics, MR766lp showed peaks of viral loads in 24 and 48 hpi for all cell types tested, as well as marked cells death after peak production. On the other hand, the HTR-8 lineage inoculated with ZIKV-IEC exhibited increased viral production in 144 hpi, with a peak between 24 and 96 hpi. Furthermore, IEC had peak variations of viral production for BeWo in 144 hpi. Considering such in vitro results, the hypothesis that maternal fetal transmission is probably a way of virus transmission between the mother and the embryo/fetus is maintained.
Collapse
Affiliation(s)
- Márcia Duarte Barbosa
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-000, Brazil
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Anderson Costa
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Paula Prieto-Oliveira
- Department of Bioinformatics and Genomics, College of Computing and Informatics, University of North Carolina at Charlotte, 9331 Robert D. Snyder Rd., Charlotte, NC 28223, USA
| | - Robert Andreata-Santos
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Cristina M Peter
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| | - Paolo M A Zanotto
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Institute of Biosciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Luiz Mario Ramos Janini
- Laboratory of Retrovirology, Department of Microbiology, Immunology and Parasitology, Discipline of Microbiology, Federal University of São Paulo, São Paulo 04039-032, Brazil
| |
Collapse
|