1
|
Song BH, Yun SI, Goldhardt JL, Kim J, Lee YM. Key virulence factors responsible for differences in pathogenicity between clinically proven live-attenuated Japanese encephalitis vaccine SA14-14-2 and its pre-attenuated highly virulent parent SA14. PLoS Pathog 2025; 21:e1012844. [PMID: 39775684 DOI: 10.1371/journal.ppat.1012844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Japanese encephalitis virus (JEV), a neuroinvasive and neurovirulent orthoflavivirus, can be prevented in humans with the SA14-14-2 vaccine, a live-attenuated version derived from the wild-type SA14 strain. To determine the viral factors responsible for the differences in pathogenicity between SA14 and SA14-14-2, we initially established a reverse genetics system that includes a pair of full-length infectious cDNAs for both strains. Using this cDNA pair, we then systematically exchanged genomic regions between SA14 and SA14-14-2 to generate 20 chimeric viruses and evaluated their replication capability in cell culture and their pathogenic potential in mice. Our findings revealed the following: (i) The single envelope (E) protein of SA14-14-2, which contains nine mutations (eight in the ectodomain and one in the stem region), is both necessary and sufficient to render SA14 non-neuroinvasive and non-neurovirulent. (ii) Conversely, the E protein of SA14 alone is necessary for SA14-14-2 to become highly neurovirulent, but it is not sufficient to make it highly neuroinvasive. (iii) The limited neuroinvasiveness of an SA14-14-2 derivative that contains the E gene of SA14 significantly increases (approaching that of the wild-type strain) when two viral nonstructural proteins are replaced by their counterparts from SA14: (a) NS1/1', which has four mutations on the external surface of the core β-ladder domain; and (b) NS2A, which has two mutations in the N-terminal region, including two non-transmembrane α-helices. In line with their roles in viral pathogenicity, the E, NS1/1', and NS2A genes all contribute to the enhanced spread of the virus in cell culture. Collectively, our data reveal for the first time that the E protein of JEV has a dual function: It is the master regulator of viral neurovirulence and also the primary initiator of viral neuroinvasion. After the initial E-mediated neuroinvasion, the NS1/1' and NS2A proteins act as secondary promoters, further amplifying viral neuroinvasiveness.
Collapse
Affiliation(s)
- Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Joseph L Goldhardt
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Jiyoun Kim
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, Utah, United States of America
| |
Collapse
|
2
|
Nanda JD, Yeh TM, Satria RD, Jhan MK, Wang YT, Lin YL, Sufriyana H, Su ECY, Lin CF, Ho TS. Dengue virus non-structural protein 1 binding to thrombin as a dengue severity marker: Comprehensive patient analysis in south Taiwan. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2024:S1684-1182(24)00230-5. [PMID: 39730269 DOI: 10.1016/j.jmii.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 07/26/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Previously we identified a complex of non-structural protein (NS) 1 - Thrombin (NST) in dengue infected patients. Here, we investigated how the concentration of NS1 and NST differ in various dengue severity levels as well as their demographic and clinical features. Several comorbid (hypertension, diabetes, and chronic renal failure) often found in dengue patients were also measured and analyzed. METHODS A total of 86 dengue patients (52 not severe and 34 severe), were enrolled and had their blood taken. Blood samples were further verified for clinical blood parameters, including liver and renal function tests and serologic assays (NS1 and NST). Patients' severity was grouped based on WHO 2009 classification, which separates patients into dengue without warning signs (DNWS), dengue with warning signs (DWWS), and severe dengue (SD). DWWS is explained as DNWS with warning signs (persistent abdominal pain, persistent vomiting, liver enlargement, bleeding (any kind), fatigue, and restlessness). SD are those with severe plasma leakage, severe bleeding, or severe organ impairment. Multivariate regression analysis was used to predict the role of NST on the dengue severity development and receiver operating characteristic (AUROC) test was utilized to evaluate separability. RESULTS The analysis revealed that NS1 significantly impacts the disease outcome (p 0.018, OR = 2.467 (1.171-5.197)) but not beyond the effect through NST (p 0.108, OR = 0.085 (0.004-1.719)). We also prove that NST was a better severity biomarker compared to NS1, as it can predict progression from DNWS to DWWS (AUC: NS1 = 0.771∗∗, NST = 0.81∗∗) and SD (AUC: NS1 = 0.607, NST = 0.754∗) significantly. CONCLUSIONS This finding suggests the importance of NST in mediating the NS1 effect to promote dengue severity progression and its promising capability as an acute stage dengue severity biomarker.
Collapse
Affiliation(s)
- Josephine Diony Nanda
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Departement of Parasitology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Rahmat Dani Satria
- Department of Clinical Pathology and Laboratory Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia; Clinical Laboratory Installation, Dr. Sardjito Central General Hospital, Yogyakarta, 55281, Indonesia
| | - Ming-Kai Jhan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Yung-Ting Wang
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Ya-Lan Lin
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan
| | - Herdiantri Sufriyana
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Department of Medical Physiology, Faculty of Medicine, Universitas Nahdlatul Ulama Surabaya, Surabaya, Indonesia
| | - Emily Chia-Yu Su
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Clinical Big Data Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan; Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei, 110, Taiwan.
| | - Tzong-Shiann Ho
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan; Department of Pediatrics, National Cheng Kung University Hospital Dou-Liou Branch, College of Medicine, National Cheng Kung University, Yunlin 640, Taiwan.
| |
Collapse
|
3
|
Das S, Mallik MH, Chattopadyay P, Mallick S, Karmakar D, Ghora S, Begum F, Chatterjee B, Thagriki DS, Srivastava AK, Ray U. Dengue virus NS1 leads to downregulation of HNF4 alpha in liver cells resulting in a decrease in coagulation factors I, V, X, and XIII, contributing to coagulopathy. J Virol 2024; 98:e0141824. [PMID: 39513713 DOI: 10.1128/jvi.01418-24] [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: 08/20/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
Dengue virus NS1 protein is a major pathogenic protein. In this study, we examined the role of NS1 in coagulopathy associated with Dengue infection, a common feature of Dengue virus pathogenesis. Since most coagulation factors are produced by hepatocytes and liver is key organ affected during infection, we conducted transcriptomics using total-RNA extracted from Huh7 cells overexpressing NS1 protein. Coagulation factors 1, 5, 10, and 13 were downregulated and was confirmed using quantitative real-time polymerase chain reaction (RT-PCR) and western blot assays in both adherent and non-adherent cell culture systems across all four serotypes of Dengue. We also determined that downregulation of coagulation factors is a result of reduced expression of transcription activator HNF4α. Furthermore, we demonstrated that phosphorylation of extracellular signal-regulated kinase (ERK) leads to HNF4α downregulation and subsequent downregulation of coagulation factors. The downregulation of HNF4α and the downregulation of subsequent coagulation factors were validated in BALB/c mice by hydrodynamic tail vein injection of NS1 expression plasmids. Western blot assays using plasma from Dengue patients indicated that at least two coagulation factors of the common pathway of coagulation cascade are downregulated during the febrile phase, with levels improving toward the convalescent phase. NS1-mediated downregulation of coagulation factors was observed for both intracellular and secreted NS1. The hypothesis was also validated using virus infection assays. Overall, our study highlights the role of NS1 in mediating coagulopathy by modulating the expression of coagulation factors through transcriptional suppression of HNF4α by elevated phosphorylated ERK. This signaling cascade could be targeted for therapeutic intervention against virus-related coagulopathies. IMPORTANCE Thrombocytopenia has been linked to coagulopathy of Dengue infection, and Dengue patients with coagulopathies are often administered platelet transfusion. For coagulopathies without thrombocytopenia, platelet transfusion might not help. We demonstrated the role of NS1 in coagulopathy by downregulating coagulation factors themselves. When thrombocytopenia does not exist or when thrombocytopenia as well as reduced levels of coagulation factors are the causative factors for coagulopathies, only platelet transfusion might not be effective. Alternative strategies, like administration of coagulation factor cocktails or platelet transfusion along with coagulation factor cocktail, might be promising. Our work also leads to a signaling pathway of NS1-mediated downregulation of coagulation factors via phosphorylated ERK and HNF4α. HNF4α is a transcription regulator for many other liver-based metabolic factors and pathways like lipid metabolism, carbohydrate metabolism, etc, and thus, therapeutic targeting of NS1-based downregulation of HNF4α can lead to designing therapeutic candidates for managing other Dengue-based liver dysfunction.
Collapse
Affiliation(s)
- Sandeepan Das
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Md Hasan Mallik
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | | | | | | | - Subhadip Ghora
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Feroza Begum
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Bilash Chatterjee
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Dluya Samuel Thagriki
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
- Adamawa State University Mubi, Nigeria, Adamawa State, Africa
| | - Amit Kumar Srivastava
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Upasana Ray
- CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
4
|
Tan H, Zhang S, Wu Z, He Y, Wang T, Tan W, Tang X, Li W, Wang M, Jia R, Zhu D, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Sun D, Tian B, Cheng A, Chen S. The greasy finger region of DTMUV NS1 plays an essential role in NS1 secretion and viral proliferation. Poult Sci 2024; 103:104322. [PMID: 39316982 PMCID: PMC11462342 DOI: 10.1016/j.psj.2024.104322] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/26/2024] Open
Abstract
Duck Tembusu virus (DTMUV) of the Orthoflavivirus genus poses a significant threat to waterfowl aquaculture. Nonstructural protein 1 (NS1), a multifunctional glycoprotein, exists in various oligomeric forms and performs diverse functions. The greasy finger (GF) region within NS1 of other flaviviruses has been shown to be a crucial component of the hydrophobic protrusion aiding in anchoring NS1 to the endoplasmic reticulum (ER). However, detailed studies on the role of the GF region in viral proliferation in vitro and the biological properties of NS1 remain scarce. A series of recombinant DTMUV (rDTMUV) with mutations in the GF region, including NS1-F158A, G159A, F160A, G161A, V162A, L163A, F160R, multipoint mutations (GF-4M), or regional deletions (ΔGF), were rescued using a DNA-based reverse genetics system. Only 5 rDTMUV variants (G159A, F160A, G161A, V162A, and L163A) could be rescued successfully, and these mutations were found to impair replication, reduce virulence, and decrease plaque size, as shown by growth kinetics, duck embryo virulence, and plaque assays, respectively. Upon examining NS1 expression by western blot, we discovered that secreted NS1 (sNS1) presented in large quantities in the supernatant of cells infected with rDTMUV-NS1-G159A, whereas intracellular NS1 was less abundant. These mutations also impacted the primary forms and secretion rates of NS1 in cases of overexpression by western blot and indirect ELISA. Exception for F160A and G161A, which showed decreased secretion rates, all other mutations increased sNS1 expression, with the most pronounced increase observed in F158A and ΔGF, and rDTMUV with these mutations can't be rescued. Co-localization studies of NS1 with the ER demonstrated that the ΔGF mutation attenuated NS1 anchoring to the ER, thereby inhibiting its intracellular residence and promoting secretion. Although these effects vary between flaviviruses, our data reveal that the GF region of NS1 is crucial for viral proliferation and NS1 secretion.
Collapse
Affiliation(s)
- Hantai Tan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Senzhao Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Zhen Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Yu He
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Tao Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Wangyang Tan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xuedan Tang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Wei Li
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Agricultural Bioinformatics, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Di Sun
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Bin Tian
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Agricultural Bioinformatics, Ministry of Education of the People's Republic of China, Chengdu 611130, China.
| |
Collapse
|
5
|
Puerta-Guardo H, Biering SB, Castillo-Rojas B, DiBiasio-White MJ, Lo NT, Espinosa DA, Warnes CM, Wang C, Cao T, Glasner DR, Beatty PR, Kuhn RJ, Harris E. Flavivirus NS1-triggered endothelial dysfunction promotes virus dissemination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.29.625931. [PMID: 39651279 PMCID: PMC11623691 DOI: 10.1101/2024.11.29.625931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The Flaviviridae are a family of viruses that include the important arthropod-borne human pathogens dengue virus (DENV), West Nile virus, Zika virus, Japanese encephalitis virus, and yellow fever virus. Flavivirus nonstructural protein 1 (NS1) is essential for virus replication but is also secreted from virus-infected cells. Extracellular NS1 acts as a virulence factor during flavivirus infection in multiple ways, including triggering endothelial dysfunction and vascular leak via interaction with endothelial cells. While the role of NS1 in inducing vascular leak and exacerbating pathogenesis is well appreciated, if and how NS1-triggered endothelial dysfunction promotes virus infection remains obscure. Flaviviruses have a common need to disseminate from circulation into specific tissues where virus-permissive cells reside. Tissue-specific dissemination is associated with disease manifestations of a given flavivirus, but mechanisms dictating virus dissemination are unclear. Here we show that NS1-mediated endothelial dysfunction promotes virus dissemination in vitro and in vivo . In mouse models of DENV infection, we show that anti-NS1 antibodies decrease virus dissemination, while the addition of exogenous NS1 promotes virus dissemination. Using an in vitro system, we show that NS1 promotes virus dissemination in two distinct ways: (1) promoting crossing of barriers and (2) increasing infectivity of target cells in a tissue- and virus-specific manner. The capacity of NS1 to modulate infectivity correlates with a physical association between virions and NS1, suggesting a potential NS1-virion interaction. Taken together, our study indicates that flavivirus NS1 promotes virus dissemination across endothelial barriers, providing an evolutionary basis for virus-triggered vascular leak. Author Summary The Flaviviridae contain numerous medically important human pathogens that cause potentially life-threatening infections. Over half of the world's population is at risk of flavivirus infection, and this number is expected to increase as climate change expands the habitats of the arthropod vectors that transmit these flaviviruses. There are few effective vaccines and no therapeutics approved for prevention or treatment of flavivirus infection, respectively. Given these challenges, understanding how and why flaviviruses cause pathogenesis is critical for identifying targets for therapeutic intervention. The secreted nonstructural protein 1 (NS1) of flaviviruses is a conserved virulence factor that triggers endothelial dysfunction in a tissue-specific manner. It is unknown if this endothelial dysfunction provides any benefit for virus infection. Here we provide evidence that NS1-triggered endothelial dysfunction facilitates virus crossing of endothelial barriers and augments infection of target cells in vitro and promotes virus dissemination in vivo . This study provides an evolutionary explanation for flaviviruses evolving the capacity to trigger barrier dysfunction and highlights NS1 and the pathways governing endothelial dysfunction, as therapeutic targets to prevent flavivirus dissemination.
Collapse
|
6
|
Apoorva, Kumar A, Singh SK. Dengue virus NS1 hits hard at the barrier integrity of human cerebral microvascular endothelial cells via cellular microRNA dysregulations. Tissue Barriers 2024:2424628. [PMID: 39508307 DOI: 10.1080/21688370.2024.2424628] [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: 08/12/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024] Open
Abstract
Dengue virus (DENV) infections are commonly reported in the tropical and subtropical regions of the world. DENV is reported to exploit various strategies to cross the blood-brain barrier. The NS1 protein of DENV plays an important role in viral neuropathogenesis, resulting in endothelial hyperpermeability and cytokine-induced vascular leak. miRNAs are short non-coding RNAs that play an important role in post-transcriptional gene regulations. However, no comprehensive information about the involvement of miRNAs in DENV-NS1-mediated neuropathogenesis has been explored to date. We observed that DENV-NS1 significantly alters the cellular miRNome of human cerebral microvascular endothelial cells in a bystander fashion. Subsequent target prediction and pathway enrichment analysis indicated that these microRNAs and their corresponding target genes are involved in pathways associated with blood-brain barrier dysfunction such as "Adherens junction" and "Tight junction". Additionally, several miRNA-mRNA pairs were also found to be involved in cellular signaling pathways related to cytokine production, for instance, "Jak-STAT signaling pathway", "Chemokine signaling pathway", "IL-17 signaling pathway", "NF-κB signaling pathway", and "Viral protein interaction with cytokine and cytokine receptor". The dysregulated production of inflammatory cytokines is reported to compromise BBB permeability. This study is the first report to demonstrate that DENV-NS1-mediated miRNA perturbations are crucial in compromising endothelial barrier integrity. It also offers insights into potential therapeutic targets to mitigate DENV-NS1-induced vascular permeability and inflammation.
Collapse
Affiliation(s)
- Apoorva
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Atul Kumar
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunit K Singh
- Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
- Dr. B.R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, New Delhi, India
- Delhi School of Public Health, University of Delhi, New Delhi, India
| |
Collapse
|
7
|
Bernardo-Menezes LC, Agrelli A, Oliveira ASLED, Azevedo EDAN, Morais CNLD. Zika virus: Critical crosstalk between pathogenesis, cytopathic effects, and macroautophagy. J Cell Biochem 2024; 125:e30438. [PMID: 37334850 DOI: 10.1002/jcb.30438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/06/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023]
Abstract
Zika virus (ZIKV) is a re-emerging positive-sense RNA arbovirus. Its genome encodes a polyprotein that is cleaved by proteases into three structural proteins (Envelope, pre-Membrane, and Capsid) and seven nonstructural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). These proteins have essential functions in viral replication cycle, cytopathic effects, and host cellular response. When infected by ZIKV, host cells promote macroautophagy, which is believed to favor virus entry. Although several authors have attempted to understand this link between macroautophagy and viral infection, little is known. Herein, we performed a narrative review of the molecular connection between macroautophagy and ZIKV infection while focusing on the roles of the structural and nonstructural proteins. We concluded that ZIKV proteins are major virulence factors that modulate host-cell machinery to its advantage by disrupting and/or blocking specific cellular systems and organelles' function, such as endoplasmic reticulum stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lucas Coêlho Bernardo-Menezes
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Almerinda Agrelli
- Laboratory of Nanostructured Materials (LMNANO), Strategic Technologies Center of Northeast (CETENE), Recife, Pernambuco, Brazil
| | | | - Elisa de Almeida Neves Azevedo
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Clarice Neuenschwander Lins de Morais
- Laboratory of Virology and Experimental Therapeutics (LaViTE), Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| |
Collapse
|
8
|
Wilken L, Rimmelzwaan GF, Elbahesh H. The Raf kinase inhibitors Dabrafenib and Regorafenib impair Zika virus replication via distinct mechanisms. J Virol 2024; 98:e0061824. [PMID: 39023323 PMCID: PMC11334485 DOI: 10.1128/jvi.00618-24] [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: 04/15/2024] [Accepted: 06/01/2024] [Indexed: 07/20/2024] Open
Abstract
Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that has been associated with congenital neurological defects in fetuses born to infected mothers. At present, no vaccine or antiviral therapy is available to combat this devastating disease. Repurposing drugs that target essential host factors exploited by viruses is an attractive therapeutic approach. Here, we screened a panel of clinically approved small-molecule kinase inhibitors for their antiviral effects against a clinical isolate of ZIKV and thoroughly characterized their mechanisms of action. We found that the Raf kinase inhibitors Dabrafenib and Regorafenib potently impair the replication of ZIKV, but not that of its close relative dengue virus. Time-of-addition experiments showed that both inhibitors target ZIKV infection at post-entry steps. We found that Dabrafenib, but not Regorafenib, interfered with ZIKV genome replication by impairing both negative- and positive-strand RNA synthesis. Regorafenib, on the other hand, altered steady-state viral protein levels, viral egress, and blocked NS1 secretion. We also observed Regorafenib-induced ER fragmentation in ZIKV-infected cells, which might contribute to its antiviral effects. Because these inhibitors target different steps of the ZIKV infection cycle, their use in combination therapy may amplify their antiviral effects which could be further explored for future therapeutic strategies against ZIKV and possibly other flaviviruses. IMPORTANCE There is an urgent need to develop effective therapeutics against re-emerging arboviruses associated with neurological disorders like Zika virus (ZIKV). We identified two FDA-approved kinase inhibitors, Dabrafenib and Regorafenib, as potent inhibitors of contemporary ZIKV strains at distinct stages of infection despite overlapping host targets. Both inhibitors reduced viral titers by ~1 to 2 log10 (~10-fold to 100-fold) with minimal cytotoxicity. Furthermore, we show that Dabrafenib inhibits ZIKV RNA replication whereas Regorafenib inhibits ZIKV translation and egress. Regorafenib has the added benefit of limiting NS1 secretion, which contributes to the pathogenesis and disease progression of several flaviviruses. Because these inhibitors affect distinct post-entry steps of ZIKV infection, their therapeutic potential may be amplified by combination therapy and likely does not require prophylactic administration. This study provides further insight into ZIKV-host interactions and has implications for the development of novel antivirals against ZIKV and possibly other flaviviruses.
Collapse
Affiliation(s)
- Lucas Wilken
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Hannover, Germany
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Hannover, Germany
| | - Husni Elbahesh
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), Hannover, Germany
| |
Collapse
|
9
|
Roy A, Paul I, Paul T, Hazarika K, Dihidar A, Ray S. An in-silico receptor-pharmacophore based multistep molecular docking and simulation study to evaluate the inhibitory potentials against NS1 of DENV-2. J Biomol Struct Dyn 2024; 42:6136-6164. [PMID: 37517062 DOI: 10.1080/07391102.2023.2239925] [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: 04/01/2023] [Accepted: 06/25/2023] [Indexed: 08/01/2023]
Abstract
DENV-2 strain is the most fatal and infectious of the five dengue virus serotypes. The non-structural protein NS1 encoded by its genome is the most significant protein required for viral pathogenesis and replication inside the host body. Thus, targeting the NS1 protein and designing an inhibitor to limit its stability and secretion is a propitious attempt in our fight against dengue. Four novel inhibitors are designed to target the conserved cysteine residues (C55, C313, C316, and C329) and glycosylation sites (N130 and N207) of the NS1 protein in an attempt to halt the spread of the dengue infection in the host body altogether. Numerous computer-aided drug designing techniques including molecular docking, molecular dynamics simulation, virtual screening, principal component analysis, and dynamic cross-correlation matrix were employed to determine the structural and functional activity of the NS1-inhibitor complexes. From our analysis, it was evident that the extent of structural and atomic level fluctuations of the ligand-bound protein exhibited a declining trend in contrast to unbound protein which was prominently noticeable through the RMSD, RMSF, Rg, and SASA graphs. The ADMET analysis of the four ligands revealed a promising pharmacokinetics and pharmacodynamic profile, along with good bioavailability and toxicity properties. The proposed drugs when bound to the targeted cavities resulted in stable conformations in comparison to their unbound state, implying they have good affinity promising effective drug action. Thus, they can be tested in vitro and used as potential anti-dengue drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alankar Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Ishani Paul
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Tanwi Paul
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | | | - Aritrika Dihidar
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Sujay Ray
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| |
Collapse
|
10
|
Rani A, Stadler JT, Marsche G. HDL-based therapeutics: A promising frontier in combating viral and bacterial infections. Pharmacol Ther 2024; 260:108684. [PMID: 38964560 DOI: 10.1016/j.pharmthera.2024.108684] [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: 02/26/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Low levels of high-density lipoprotein (HDL) and impaired HDL functionality have been consistently associated with increased susceptibility to infection and its serious consequences. This has been attributed to the critical role of HDL in maintaining cellular lipid homeostasis, which is essential for the proper functioning of immune and structural cells. HDL, a multifunctional particle, exerts pleiotropic effects in host defense against pathogens. It functions as a natural nanoparticle, capable of sequestering and neutralizing potentially harmful substances like bacterial lipopolysaccharides. HDL possesses antiviral activity, preventing viruses from entering or fusing with host cells, thereby halting their replication cycle. Understanding the complex relationship between HDL and the immune system may reveal innovative targets for developing new treatments to combat infectious diseases and improve patient outcomes. This review aims to emphasize the role of HDL in influencing the course of bacterial and viral infections and its and its therapeutic potential.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Styria, Austria.
| |
Collapse
|
11
|
Pan Q, Chen Q, Zhang W, Jiao H, Yu L, Hu H. Structural Insights into the Dynamic Assembly of a YFV sNS1 Tetramer. Viruses 2024; 16:1212. [PMID: 39205186 PMCID: PMC11359903 DOI: 10.3390/v16081212] [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: 06/05/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 09/04/2024] Open
Abstract
Yellow fever virus (YFV) infections can cause severe diseases in humans, resulting in mass casualties in Africa and the Americas each year. Secretory NS1 (sNS1) is thought to be used as a diagnostic marker of flavivirus infections, playing an essential role in the flavivirus life cycle, but little is known about the composition and structure of YFV sNS1. Here, we present that the recombinant YFV sNS1 exists in a heterogeneous mixture of oligomerizations, predominantly in the tetrameric form. The cryoEM structures show that the YFV tetramer of sNS1 is stacked by the hydrophobic interaction between β-roll domains and greasy fingers. According to the 3D variability analysis, the tetramer is in a semi-stable state that may contain multiple conformations with dynamic changes. We believe that our study provides critical insights into the oligomerization of NS1 and will aid the development of NS1-based diagnoses and therapies.
Collapse
Affiliation(s)
- Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Wanqin Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Lei Yu
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| |
Collapse
|
12
|
Alvin Chew BL, Pan Q, Hu H, Luo D. Structural biology of flavivirus NS1 protein and its antibody complexes. Antiviral Res 2024; 227:105915. [PMID: 38777094 DOI: 10.1016/j.antiviral.2024.105915] [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: 01/30/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
The genus of flavivirus includes many mosquito-borne human pathogens, such as Zika (ZIKV) and the four serotypes of dengue (DENV1-4) viruses, that affect billions of people as evidenced by epidemics and endemicity in many countries and regions in the world. Among the 10 viral proteins encoded by the viral genome, the nonstructural protein 1 (NS1) is the only secreted protein and has been used as a diagnostic biomarker. NS1 has also been an attractive target for its biotherapeutic potential as a vaccine antigen. This review focuses on the recent advances in the structural landscape of the secreted NS1 (sNS1) and its complex with monoclonal antibodies (mAbs). NS1 forms an obligatory dimer, and upon secretion, it has been reported to be hexametric (trimeric dimers) that could dissociate and bind to the epithelial cell membrane. However, high-resolution structural information has been missing about the high-order oligomeric states of sNS1. Several cryoEM studies have since shown that DENV and ZIKV recombinant sNS1 (rsNS1) are in dynamic equilibrium of dimer-tetramer-hexamer states, with tetramer being the predominant form. It was recently revealed that infection-derived sNS1 (isNS1) forms a complex of the NS1 dimer partially embedded in a High-Density Lipoprotein (HDL) particle. Structures of NS1 in complexes with mAbs have also been reported which shed light on their protective roles during infection. The biological significance of the diversity of NS1 oligomeric states remains to be further studied, to inform future research on flaviviral pathogenesis and the development of therapeutics and vaccines. Given the polymorphism of flavivirus NS1 across sample types with variations in antigenicity, we propose a nomenclature to accurately define NS1 based on the localization and origin.
Collapse
Affiliation(s)
- Bing Liang Alvin Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore, 636921; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore, 636921.
| | - Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore, 636921; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore, 636921; National Centre for Infectious Diseases, Singapore, 308442, Singapore.
| |
Collapse
|
13
|
Muthukumaran R, Sankararamakrishnan R. Differences in the Membrane-Binding Properties of Flaviviral Nonstructural 1 (NS1) Protein: Comparative Simulations of Zika and Dengue Virus NS1 Proteins in Explicit Bilayers. ACS BIO & MED CHEM AU 2024; 4:137-153. [PMID: 38911907 PMCID: PMC11191575 DOI: 10.1021/acsbiomedchemau.3c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 06/25/2024]
Abstract
NS1 in flaviviruses is the only nonstructural protein that is secretory and interacts with different cellular components of the host cell membrane. NS1 is localized in the ER as a dimer to facilitate viral replication. Crystal structures of NS1 homologues from zika (ZIKV) and dengue (DENV) viruses have revealed the organization of different domains in NS1 dimers. The β-roll and the connector and intertwined loop regions of wing domains of NS1 have been shown to interact with the membranes. In this study, we have performed multiple molecular dynamics (MD) simulations of ZIKV and DENV NS1 systems in apo and in POPE bilayers with different cholesterol concentrations (0, 20 and 40%). The NS1 protein was placed just above the membrane surface, and for each NS1-membrane system two to three independent simulations with 600 ns production run were performed. At the end of the production runs, ZIKV NS1 inserts deeper inside the membrane compared to the DENV counterpart. Unlike ZIKV NS1, the orientation of DENV NS1 is asymmetric in which one of the chains in the dimer interacts with the membrane while the other is more exposed to the solvent. The β-roll region in ZIKV NS1 penetrates beyond the headgroup region and interacts with the lipid acyl chains while the C-terminal region barely interacts with the headgroup. Specific residues in the intertwined region deeply penetrate inside the membrane. The role of charged and aromatic residues of ZIKV NS1 in strongly interacting with the membrane components is revealed. The presence of cholesterol affects the extent of insertion in the membrane and interaction of individual residues. Overall, membrane-binding properties of ZIKV NS1 significantly differ from its counterpart in DENV. The differences found in the binding and insertion of NS1 can be used to design drugs and novel antibodies that can be flavivirus specific.
Collapse
Affiliation(s)
- Rajagopalan Muthukumaran
- Department
of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ramasubbu Sankararamakrishnan
- Department
of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
- Mehta
Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
14
|
Sitthisuwannakul K, Sukthai R, Zhu Z, Nagashima K, Chattrairat K, Phanthanawiboon S, Klamchuen A, Rahong S, Baba Y, Yasui T. Urinary dengue NS1 detection on Au-decorated ZnO nanowire platform. Biosens Bioelectron 2024; 254:116218. [PMID: 38518559 DOI: 10.1016/j.bios.2024.116218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/17/2024] [Accepted: 03/12/2024] [Indexed: 03/24/2024]
Abstract
Biodetection for non-invasive diagnostics of fluids, especially urine, remains a challenge to scientists due to low target concentrations. And biological complexes of the detection target may contain contaminants that also interfere with any assay. Dengue non-structural 1 protein (Dengue NS1) is an important biomarker for dengue hemorrhagic fever and dengue shock syndrome. Here, we developed an Au-decorated nanowire platform and applied it with a sandwich fluorophore-linked immunosorbent well plate assay (FLISA) to detect Dengue NS1 in urine. For the platform, we fabricated zinc oxide (ZnO) nanowires to provide a high surface area and then coated them with gold nanoparticles (ZnO/Au nanowires) to simply modify the Dengue NS1 antibody and enhance the fluorescence intensity. Our platform employs a sandwich FLISA that exhibits high sensitivity, specifically detecting Dengue NS1 with a limit of detection (LOD) of 1.35 pg/mL. This LOD was 4500-fold lower than the LOD of a commercially available kit for Dengue NS1 enzyme-linked immunosorbent assay. We believe that our ZnO/Au nanowire platform has the potential to revolutionize the field of non-invasive diagnostics for dengue.
Collapse
Affiliation(s)
- Kannika Sitthisuwannakul
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta 4259, Midori-ku, Yokohama, 226-8501, Japan.
| | - Ratchanon Sukthai
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Zetao Zhu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta 4259, Midori-ku, Yokohama, 226-8501, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Kazuki Nagashima
- Research Institute for Electronic Science (RIES), Hokkaido University, N21W10, Kita, Sapporo, Hokkaido, 001-0021, Japan
| | - Kunanon Chattrairat
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta 4259, Midori-ku, Yokohama, 226-8501, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | | | - Annop Klamchuen
- National Nanotechnology Center (NANOTEC), NSTDA, Pathum Thani, 12120, Thailand
| | - Sakon Rahong
- College of Materials Innovation and Technology, King Mongkut's Institute of Technology Ladkrabang, Chalongkrung Rd., Ladkrabang, Bangkok, 10520, Thailand
| | - Yoshinobu Baba
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Institute of Quantum Life Science, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan.
| | - Takao Yasui
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Department of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta 4259, Midori-ku, Yokohama, 226-8501, Japan; Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan; Institute of Quantum Life Science, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba, 263-8555, Japan.
| |
Collapse
|
15
|
Chew BLA, Ngoh ANQ, Phoo WW, Chan KWK, Ser Z, Tulsian NK, Lim SS, Weng MJG, Watanabe S, Choy MM, Low J, Ooi EE, Ruedl C, Sobota RM, Vasudevan SG, Luo D. Secreted dengue virus NS1 from infection is predominantly dimeric and in complex with high-density lipoprotein. eLife 2024; 12:RP90762. [PMID: 38787378 PMCID: PMC11126310 DOI: 10.7554/elife.90762] [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] [Indexed: 05/25/2024] Open
Abstract
Severe dengue infections are characterized by endothelial dysfunction shown to be associated with the secreted nonstructural protein 1 (sNS1), making it an attractive vaccine antigen and biotherapeutic target. To uncover the biologically relevant structure of sNS1, we obtained infection-derived sNS1 (isNS1) from dengue virus (DENV)-infected Vero cells through immunoaffinity purification instead of recombinant sNS1 (rsNS1) overexpressed in insect or mammalian cell lines. We found that isNS1 appeared as an approximately 250 kDa complex of NS1 and ApoA1 and further determined the cryoEM structures of isNS1 and its complex with a monoclonal antibody/Fab. Indeed, we found that the major species of isNS1 is a complex of the NS1 dimer partially embedded in a high-density lipoprotein (HDL) particle. Crosslinking mass spectrometry studies confirmed that the isNS1 interacts with the major HDL component ApoA1 through interactions that map to the NS1 wing and hydrophobic domains. Furthermore, our studies demonstrated that the sNS1 in sera from DENV-infected mice and a human patient form a similar complex as isNS1. Our results report the molecular architecture of a biological form of sNS1, which may have implications for the molecular pathogenesis of dengue.
Collapse
Affiliation(s)
- Bing Liang Alvin Chew
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| | - AN Qi Ngoh
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Wint Wint Phoo
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Kitti Wing Ki Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Zheng Ser
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Nikhil K Tulsian
- Department of Biological Sciences, National University of SingaporeSingaporeSingapore
- Singapore Centre for Life Sciences, Department of Biochemistry, National University of SingaporeSingaporeSingapore
| | - Shiao See Lim
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Mei Jie Grace Weng
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| | - Satoru Watanabe
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Milly M Choy
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
| | - Jenny Low
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Department of Infectious Diseases, Singapore General HospitalSingaporeSingapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Saw Swee Hock School of Public Health, National University of SingaporeSingaporeSingapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological UniversitySingaporeSingapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingaporeSingapore
| | - Subhash G Vasudevan
- Program in Emerging Infectious Diseases, Duke-NUS Medical SchoolSingaporeSingapore
- Department of Microbiology and Immunology, National University of SingaporeSingaporeSingapore
- Institute for Glycomics (G26), Griffith University Gold Coast CampusSouthportAustralia
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- NTU Institute of Structural Biology, Nanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
16
|
Sornjai W, Promma P, Priewkhiew S, Ramphan S, Jaratsittisin J, Jinagool P, Wikan N, Greenwood M, Murphy D, Smith DR. The interaction of GRP78 and Zika virus E and NS1 proteins occurs in a chaperone-client manner. Sci Rep 2024; 14:10407. [PMID: 38710792 PMCID: PMC11074156 DOI: 10.1038/s41598-024-61195-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: 01/03/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
Glucose regulated protein 78 (GRP78) is a chaperone protein that is a central mediator of the unfolded protein response, a key cellular stress response pathway. GRP78 has been shown to be critically required for infection and replication of a number of flaviviruses, and to interact with both non-structural (NS) and structural flavivirus proteins. However, the nature of the specific interaction between GRP78 and viral proteins remains largely unknown. This study aimed to characterize the binding domain and critical amino acid residues that mediate the interaction of GRP78 to ZIKV E and NS1 proteins. Recombinant EGFP fused GRP78 and individual subdomains (the nucleotide binding domain (NBD) and the substrate binding domain (SBD)) were used as a bait protein and co-expressed with full length or truncated ZIKV E and NS1 proteins in HEK293T/17 cells. Protein-protein interactions were determined by a co-immunoprecipitation assay. From the results, both the NBD and the SBD of GRP78 were crucial for an effective interaction. Single amino acid substitutions in the SBD showed that R492E and T518A mutants significantly reduced the binding affinity of GRP78 to ZIKV E and NS1 proteins. Notably, the interaction of GRP78 with ZIKV E was stably maintained against various single amino acid substitutions on ZIKV E domain III and with all truncated ZIKV E and NS1 proteins. Collectively, the results suggest that the principal binding between GRP78 and viral proteins is mainly a classic canonical chaperone protein-client interaction. The blocking of GRP78 chaperone function effectively inhibited ZIKV infection and replication in neuronal progenitor cells. Our findings reveal that GRP78 is a potential host target for anti-ZIKV therapeutics.
Collapse
Affiliation(s)
- Wannapa Sornjai
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Ploenphit Promma
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Suphansa Priewkhiew
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Suwipa Ramphan
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Janejira Jaratsittisin
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Pailin Jinagool
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
| | - Nitwara Wikan
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Michael Greenwood
- Molecular Neuroendocrinology Research Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol, UK
| | - Duncan R Smith
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phutthamonthon Sai 4 Road, Salaya, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
17
|
Pan Q, Jiao H, Zhang W, Chen Q, Zhang G, Yu J, Zhao W, Hu H. The step-by-step assembly mechanism of secreted flavivirus NS1 tetramer and hexamer captured at atomic resolution. SCIENCE ADVANCES 2024; 10:eadm8275. [PMID: 38691607 PMCID: PMC11062569 DOI: 10.1126/sciadv.adm8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/01/2024] [Indexed: 05/03/2024]
Abstract
Flaviviruses encode a conserved, membrane-associated nonstructural protein 1 (NS1) with replication and immune evasion functions. The current knowledge of secreted NS1 (sNS1) oligomers is based on several low-resolution structures, thus hindering the development of drugs and vaccines against flaviviruses. Here, we revealed that recombinant sNS1 from flaviviruses exists in a dynamic equilibrium of dimer-tetramer-hexamer states. Two DENV4 hexameric NS1 structures and several tetrameric NS1 structures from multiple flaviviruses were solved at atomic resolution by cryo-EM. The stacking of the tetrameric NS1 and hexameric NS1 is facilitated by the hydrophobic β-roll and connector domains. Additionally, a triacylglycerol molecule located within the central cavity may play a role in stabilizing the hexamer. Based on differentiated interactions between the dimeric NS1, two distinct hexamer models (head-to-head and side-to-side hexamer) and the step-by-step assembly mechanisms of NS1 dimer into hexamer were proposed. We believe that our study sheds light on the understanding of the NS1 oligomerization and contributes to NS1-based therapies.
Collapse
Affiliation(s)
- Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Wanqin Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Geshu Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| | - Jianhai Yu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong 518172, China
| |
Collapse
|
18
|
Sinha S, Singh K, Ravi Kumar YS, Roy R, Phadnis S, Meena V, Bhattacharyya S, Verma B. Dengue virus pathogenesis and host molecular machineries. J Biomed Sci 2024; 31:43. [PMID: 38649998 PMCID: PMC11036733 DOI: 10.1186/s12929-024-01030-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/14/2024] [Indexed: 04/25/2024] Open
Abstract
Dengue viruses (DENV) are positive-stranded RNA viruses belonging to the Flaviviridae family. DENV is the causative agent of dengue, the most rapidly spreading viral disease transmitted by mosquitoes. Each year, millions of people contract the virus through bites from infected female mosquitoes of the Aedes species. In the majority of individuals, the infection is asymptomatic, and the immune system successfully manages to control virus replication within a few days. Symptomatic individuals may present with a mild fever (Dengue fever or DF) that may or may not progress to a more critical disease termed Dengue hemorrhagic fever (DHF) or the fatal Dengue shock syndrome (DSS). In the absence of a universally accepted prophylactic vaccine or therapeutic drug, treatment is mostly restricted to supportive measures. Similar to many other viruses that induce acute illness, DENV has developed several ways to modulate host metabolism to create an environment conducive to genome replication and the dissemination of viral progeny. To search for new therapeutic options, understanding the underlying host-virus regulatory system involved in various biological processes of the viral life cycle is essential. This review aims to summarize the complex interaction between DENV and the host cellular machinery, comprising regulatory mechanisms at various molecular levels such as epigenetic modulation of the host genome, transcription of host genes, translation of viral and host mRNAs, post-transcriptional regulation of the host transcriptome, post-translational regulation of viral proteins, and pathways involved in protein degradation.
Collapse
Affiliation(s)
- Saumya Sinha
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Kinjal Singh
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Y S Ravi Kumar
- Department of Biotechnology, M. S. Ramaiah Institute of Technology, MSR Nagar, Bengaluru, India
| | - Riya Roy
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sushant Phadnis
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Varsha Meena
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sankar Bhattacharyya
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Bhupendra Verma
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| |
Collapse
|
19
|
Pereira SH, Sá Magalhães Serafim M, Moraes TDFS, Zini N, Abrahão JS, Nogueira ML, Coelho dos Reis JGA, Bagno FF, da Fonseca FG. Design, development, and validation of multi-epitope proteins for serological diagnosis of Zika virus infections and discrimination from dengue virus seropositivity. PLoS Negl Trop Dis 2024; 18:e0012100. [PMID: 38635656 PMCID: PMC11025737 DOI: 10.1371/journal.pntd.0012100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Zika virus (ZIKV), an arbovirus from the Flaviviridae family, is the causative agent of Zika fever, a mild and frequent oligosymptomatic disease in humans. Nonetheless, on rare occasions, ZIKV infection can be associated with Guillain-Barré Syndrome (GBS), and severe congenital complications, such as microcephaly. The oligosymptomatic disease, however, presents symptoms that are quite similar to those observed in infections caused by other frequent co-circulating arboviruses, including dengue virus (DENV). Moreover, the antigenic similarity between ZIKV and DENV, and even with other members of the Flaviviridae family, complicates serological testing due to the high cross-reactivity of antibodies. Here, we designed, produced in a prokaryotic expression system, and purified three multiepitope proteins (ZIKV-1, ZIKV-2, and ZIKV-3) for differential diagnosis of Zika. The proteins were evaluated as antigens in ELISA tests for the detection of anti-ZIKV IgG using ZIKV- and DENV-positive human sera. The recombinant proteins were able to bind and detect anti-ZIKV antibodies without cross-reactivity with DENV-positive sera and showed no reactivity with Chikungunya virus (CHIKV)- positive sera. ZIKV-1, ZIKV-2, and ZIKV-3 proteins presented 81.6%, 95%, and 66% sensitivity and 97%, 96%, and 84% specificity, respectively. Our results demonstrate the potential of the designed and expressed antigens in the development of specific diagnostic tests for the detection of IgG antibodies against ZIKV, especially in regions with the circulation of multiple arboviruses.
Collapse
Affiliation(s)
- Samille Henriques Pereira
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mateus Sá Magalhães Serafim
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thaís de Fátima Silva Moraes
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Nathalia Zini
- Laboratório de Pesquisa em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jônatas Santos Abrahão
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maurício Lacerda Nogueira
- Laboratório de Pesquisa em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, São Paulo, Brazil
| | | | - Flávia Fonseca Bagno
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Centro de Tecnologia em Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
20
|
Wong MP, Juan EYW, Pahmeier F, Chelluri SS, Wang P, Castillo-Rojas B, Blanc SF, Biering SB, Vance RE, Harris E. The inflammasome pathway is activated by dengue virus non-structural protein 1 and is protective during dengue virus infection. PLoS Pathog 2024; 20:e1012167. [PMID: 38662771 DOI: 10.1371/journal.ppat.1012167] [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: 10/03/2023] [Revised: 05/07/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Dengue virus (DENV) is a medically important flavivirus causing an estimated 50-100 million dengue cases annually, some of whom progress to severe disease. DENV non-structural protein 1 (NS1) is secreted from infected cells and has been implicated as a major driver of dengue pathogenesis by inducing endothelial barrier dysfunction. However, less is known about how DENV NS1 interacts with immune cells and what role these interactions play. Here we report that DENV NS1 can trigger activation of inflammasomes, a family of cytosolic innate immune sensors that respond to infectious and noxious stimuli, in mouse and human macrophages. DENV NS1 induces the release of IL-1β in a caspase-1 dependent manner. Additionally, we find that DENV NS1-induced inflammasome activation is independent of the NLRP3, Pyrin, and AIM2 inflammasome pathways, but requires CD14. Intriguingly, DENV NS1-induced inflammasome activation does not induce pyroptosis and rapid cell death; instead, macrophages maintain cellular viability while releasing IL-1β. Lastly, we show that caspase-1/11-deficient, but not NLRP3-deficient, mice are more susceptible to lethal DENV infection. Together, these results indicate that the inflammasome pathway acts as a sensor of DENV NS1 and plays a protective role during infection.
Collapse
Affiliation(s)
- Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Evan Y W Juan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Sai S Chelluri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Phoebe Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, California, United States of America
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, California, United States of America
| |
Collapse
|
21
|
Hasan ME, Samir A, Khalil MM, Shafaa MW. Bioinformatics approach for prediction and analysis of the Non-Structural Protein 4B (NSP4B) of the Zika virus. J Genet Eng Biotechnol 2024; 22:100336. [PMID: 38494248 PMCID: PMC10860876 DOI: 10.1016/j.jgeb.2023.100336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
BACKGROUND The Nonstructural Protein (NSP) 4B of Zika virus of 251 amino acids from (ZIKV/Human/POLG_ZIKVF) with accession number (A0A024B7W1), Induces the production of Endoplasmic Reticulum ER-derived membrane vesicles, which are the sites of viral replication. To understand the physical basis of how proteins fold in nature and to solve the challenge of protein structure prediction, Ab-initio and comparative modeling are crucial tools. RESULTS The systematic in silico technique, ThreaDom, had only predicted one domain (4 - 190) of NSP4B. I-TASSER, and Alphafold were ranked as the best servers for full-length 3-D protein structure predictions of NSP4B, where the predicted models were evaluated quantitatively using benchmarked metrics including C-score (-3.43), TM-score (0.77949), RMSD (2.73), and Z-score (1.561). The functional and protein binding motifs were realized using motif databases, secondary and surface accessibility predictions combined with Post-Translational Modification Sites (PTMs) prediction. Two highly conserved protein-binding motifs (Flavi NS4B and Bacillus papRprotein), together with three (PTMs) (Casein Kinase II, Myristyl site, and ASN-Glycosylation site) were predicted utilizing the Motif scan and Scanprosite servers. These patterns and PTMs were associated with NSP4B's role in triggering the development of the viral replication complex and its participation in the localization of NS3 and NS5 on the membrane. Only one hit from Structural Classification of Protein (SCOP) matched the protein sequence at positions 10 to 397 and was categorized six-hairpin glycosidases superfamily according to CATH (Class, Architecture, Topology, and Homology). Integrating this NSP4B information with the templates' SCOP and CATH annotations achieves it easier to attribute structure-function/evolution links to both previously known and recently discovered protein structures.
Collapse
Affiliation(s)
- Mohamed E Hasan
- Bioinformatics Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City 32897, Egypt.
| | - Aya Samir
- Physics Department, Medical Biophysics Division, Faculty of Science, Helwan University, Cairo, Egypt
| | - Magdy M Khalil
- Physics Department, Medical Biophysics Division, Faculty of Science, Helwan University, Cairo, Egypt; School of Biotechnology, Badr University in Cairo, Egypt
| | - Medhat W Shafaa
- Physics Department, Medical Biophysics Division, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
22
|
Lim HJ, Saha T, Ooi CW. Site-specific imprinting of dengue virus non-structural 1 antigen on a polydopamine-based sensing film for early detection and prognosis of dengue. Talanta 2024; 268:125376. [PMID: 37951180 DOI: 10.1016/j.talanta.2023.125376] [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: 07/13/2023] [Revised: 10/06/2023] [Accepted: 10/30/2023] [Indexed: 11/13/2023]
Abstract
Serum levels of dengue virus (DENV) non-structural 1 (NS1) antigen can serve as a valuable prognostic indicator of severe dengue infections. A quartz crystal microbalance (QCM)-based biosensor with a biomimetic recognition element was designed to quantitatively detect DENV NS1 as an early disease biomarker. To mitigate the reliance on costly viral antigens during the molecular imprinting process, a synthetic peptide mimicking a DENV NS1 epitope was used as a surrogate template for the synthesis of an epitope-imprinted polydopamine (EMIPDA) sensing film on the biosensor surface. The maximal frequency shift for DENV NS1 was obtained with an EMIPDA film synthesised using 5 mg mL-1 of dopamine monomer and 0.5 mg mL-1 of peptide template. The EMIPDA-QCM biosensor achieved low detection and quantitation limits of 0.091 μg mL-1 and 0.436 μg mL-1, respectively, allowing acute-phase detection of dengue and prognosis of the disease progression. The EMIPDA-QCM biosensor exhibited remarkable selectivity with up to 68-fold larger frequency responses towards DENV NS1 compared to a major serum protein. The site-specific imprinting approach not only enhanced the biosensing performance but also enabled a 26-fold cost reduction for biosensor functionalisation, providing a cost-effective strategy for label-free biosensing of the dengue biomarker via the biopolymer film.
Collapse
Affiliation(s)
- Hui Jean Lim
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Tridib Saha
- Department of Electrical and Robotics Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chien Wei Ooi
- Department of Chemical Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia; Advanced Engineering Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia; Tropical Medicine and Biology Platform, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
23
|
Morales SV, Coelho GM, Ricciardi-Jorge T, Dorl GG, Zanluca C, Duarte Dos Santos CN. Development of a quantitative NS1 antigen enzyme-linked immunosorbent assay (ELISA) for Zika virus detection using a novel virus-specific mAb. Sci Rep 2024; 14:2544. [PMID: 38291109 PMCID: PMC10827715 DOI: 10.1038/s41598-024-52123-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/14/2024] [Indexed: 02/01/2024] Open
Abstract
Viruses from the Flaviviridae family, such as Dengue virus (DENV), Yellow fever virus (YFV), and Zika virus (ZIKV) are notorious global public health problems. ZIKV emergence in Polynesia and the Americas from 2013 to 2016 raised concerns as new distinguishing features set it apart from previous outbreaks, including its association with neurological complications and heightened disease severity. Virus detection is impaired as cross-reactivity to other closely related orthoflaviviruses is common among commercially available diagnostic kits. While non-structural protein 1 (NS1) has been used as an early marker of DENV and West Nile virus (WNV) infection, little is known about NS1 expression during ZIKV infection. In the present work, we developed a NS1 capture ELISA using a novel ZIKV-specific monoclonal antibody to study NS1 expression dynamics in vitro in mosquito and human cell lines. While detectable in culture supernatants, higher concentrations of NS1 were predominantly cell-associated. To our knowledge, this is the first report of NS1 detection in human cells despite viral clearance over time. Tests with human samples need to be conducted to validate the applicability of NS1 detection for diagnosis, but overall, the tools developed in this work are promising for specific detection of acute ZIKV infection.
Collapse
Affiliation(s)
| | - Gabriela Mattoso Coelho
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, FIOCRUZ, Curitiba, Paraná, Brazil
| | | | - Gisiane Gruber Dorl
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, FIOCRUZ, Curitiba, Paraná, Brazil
| | - Camila Zanluca
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, FIOCRUZ, Curitiba, Paraná, Brazil
| | | |
Collapse
|
24
|
Perera DR, Ranadeva ND, Sirisena K, Wijesinghe KJ. Roles of NS1 Protein in Flavivirus Pathogenesis. ACS Infect Dis 2024; 10:20-56. [PMID: 38110348 DOI: 10.1021/acsinfecdis.3c00566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Flaviviruses such as dengue, Zika, and West Nile viruses are highly concerning pathogens that pose significant risks to public health. The NS1 protein is conserved among flaviviruses and is synthesized as a part of the flavivirus polyprotein. It plays a critical role in viral replication, disease progression, and immune evasion. Post-translational modifications influence NS1's stability, secretion, antigenicity, and interactions with host factors. NS1 protein forms extensive interactions with host cellular proteins allowing it to affect vital processes such as RNA processing, gene expression regulation, and cellular homeostasis, which in turn influence viral replication, disease pathogenesis, and immune responses. NS1 acts as an immune evasion factor by delaying complement-dependent lysis of infected cells and contributes to disease pathogenesis by inducing endothelial cell damage and vascular leakage and triggering autoimmune responses. Anti-NS1 antibodies have been shown to cross-react with host endothelial cells and platelets, causing autoimmune destruction that is hypothesized to contribute to disease pathogenesis. However, in contrast, immunization of animal models with the NS1 protein confers protection against lethal challenges from flaviviruses such as dengue and Zika viruses. Understanding the multifaceted roles of NS1 in flavivirus pathogenesis is crucial for effective disease management and control. Therefore, further research into NS1 biology, including its host protein interactions and additional roles in disease pathology, is imperative for the development of strategies and therapeutics to combat flavivirus infections successfully. This Review provides an in-depth exploration of the current available knowledge on the multifaceted roles of the NS1 protein in the pathogenesis of flaviviruses.
Collapse
Affiliation(s)
- Dayangi R Perera
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
| | - Nadeeka D Ranadeva
- Department of Biomedical Science, Faculty of Health Sciences, KIU Campus Sri Lanka 10120
| | - Kavish Sirisena
- Department of Chemistry, Faculty of Science, University of Colombo, Sri Lanka 00300
- Section of Genetics, Institute for Research and Development in Health and Social Care, Sri Lanka 10120
| | | |
Collapse
|
25
|
Jain S, Vimal N, Angmo N, Sengupta M, Thangaraj S. Dengue Vaccination: Towards a New Dawn of Curbing Dengue Infection. Immunol Invest 2023; 52:1096-1149. [PMID: 37962036 DOI: 10.1080/08820139.2023.2280698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dengue is an infectious disease caused by dengue virus (DENV) and is a serious global burden. Antibody-dependent enhancement and the ability of DENV to infect immune cells, along with other factors, lead to fatal Dengue Haemorrhagic Fever and Dengue Shock Syndrome. This necessitates the development of a robust and efficient vaccine but vaccine development faces a number of hurdles. In this review, we look at the epidemiology, genome structure and cellular targets of DENV and elaborate upon the immune responses generated by human immune system against DENV infection. The review further sheds light on various challenges in development of a potent vaccine against DENV which is followed by presenting a current account of different vaccines which are being developed or have been licensed.
Collapse
Affiliation(s)
- Sidhant Jain
- Independent Researcher, Institute for Globally Distributed Open Research and Education (IGDORE), Rewari, India
| | - Neha Vimal
- Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Nilza Angmo
- Maitreyi College, University of Delhi, Delhi, India
| | - Madhumita Sengupta
- Janki Devi Bajaj Government Girls College, University of Kota, Kota, India
| | - Suraj Thangaraj
- Swami Ramanand Teerth Rural Government Medical College, Maharashtra University of Health Sciences, Ambajogai, India
| |
Collapse
|
26
|
Denolly S, Guo H, Martens M, Płaszczyca A, Scaturro P, Prasad V, Kongmanas K, Punyadee N, Songjaeng A, Mairiang D, Pichlmair A, Avirutnan P, Bartenschlager R. Dengue virus NS1 secretion is regulated via importin-subunit β1 controlling expression of the chaperone GRp78 and targeted by the clinical drug ivermectin. mBio 2023; 14:e0144123. [PMID: 37702492 PMCID: PMC10653883 DOI: 10.1128/mbio.01441-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 09/14/2023] Open
Abstract
IMPORTANCE Dengue virus (DENV) is a major human pathogen that can cause hemorrhagic fever and shock syndrome. One important factor of DENV pathogenicity is non-structural protein 1 (NS1), a glycoprotein that is secreted from infected cells. Here we study the mode of action of the widely used drug ivermectin, used to treat parasitic infections and recently shown to lower NS1 blood levels in DENV-infected patients. We found that ivermectin blocks the nuclear transport of transcription factors required for the expression of chaperones that support the folding and secretion of glycoproteins, including NS1. Impairing nuclear transport of these transcription factors by ivermectin or depleting them from infected cells dampens NS1 folding and thus its secretion. These results reveal a novel mode of action of ivermectin that might apply to other flaviviruses as well.
Collapse
Affiliation(s)
- Solène Denolly
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Hongbo Guo
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Miriam Martens
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Anna Płaszczyca
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Pietro Scaturro
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
- Leibniz Institute of Virology, Hamburg, Germany
| | - Vibhu Prasad
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| | - Kessiri Kongmanas
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nuntaya Punyadee
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Adisak Songjaeng
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Dumrong Mairiang
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Andreas Pichlmair
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
| | - Panisadee Avirutnan
- Division of Dengue Hemorrhagic Fever Research, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, Heidelberg University, Medical Faculty Heidelberg, Heidelberg, Germany
| |
Collapse
|
27
|
Zeng Q, Liu J, Hao C, Zhang B, Zhang H. Making sense of flavivirus non-strctural protein 1 in innate immune evasion and inducing tissue-specific damage. Virus Res 2023; 336:199222. [PMID: 37716670 PMCID: PMC10518729 DOI: 10.1016/j.virusres.2023.199222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Flaviviruses include medically important mosquito-borne pathogens, such as Zika virus (ZIKV), Japanese encephalitis virus (JEV), dengue virus (DENV) and West Nile virus (WNV), that cause hundreds of millions of infections each year. Currently, there are no approved effect therapies against mosquito-borne flaviviruses. The flaviviruses encoded nonstructural protein 1 (NS1) is a secreted glycoprotein widely involved in viral replication, immune evasion, and directly causing tissue-specific damage during flaviviruses infection. Upon viral infection of host cell, NS1 can be found in multiple oligomeric forms and include a dimer on the cell surface, and a soluble secreted hexameric lipoparticle. In the recent decade, the detailed crystal structure of several flaviviruses NS1 have been determined and unraveled its broader and deeper functions. Consistent with the potential immune function revealed by its structure, NS1 is involved in the escaping of host signal immune pathway mediated by pattern recognition receptors (PRRs), including RIG-I-like receptors (RLRS) and Toll-like receptors (TLRs). Moreover, the flavivirus NS1 is efficiently secreted by infected cells and circulates in the blood of the host to directly induce specific tissues damage. The NS1 of ZIKV, JEV and WNV changes the permeability of brain microvascular endothelial cell to cause endothelial cell dysfunction and promote virus pathogenesis. DENV NS1 can induce systemic tissues damage in humans through multiple strategies. Mutations of several key amino acids in NS1 can reduce the neurovirulence of the flavivirus. In this article, we provide an overview of the latest research on this fascinating protein in these disparate areas.
Collapse
Affiliation(s)
- Quan Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Jiaqi Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Chenlin Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Honglei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China.
| |
Collapse
|
28
|
Wong MP, Juan EYW, Chelluri SS, Wang P, Pahmeier F, Castillo-Rojas B, Blanc SF, Biering SB, Vance RE, Harris E. The Inflammasome Pathway is Activated by Dengue Virus Non-structural Protein 1 and is Protective During Dengue Virus Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558875. [PMID: 37790301 PMCID: PMC10543007 DOI: 10.1101/2023.09.21.558875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dengue virus (DENV) is a medically important flavivirus causing an estimated 50-100 million dengue cases annually, some of whom progress to severe disease. DENV non-structural protein 1 (NS1) is secreted from infected cells and has been implicated as a major driver of dengue pathogenesis by inducing endothelial barrier dysfunction. However, less is known about how DENV NS1 interacts with immune cells and what role these interactions play. Here we report that DENV NS1 can trigger activation of inflammasomes, a family of cytosolic innate immune sensors that respond to infectious and noxious stimuli, in mouse and human macrophages. DENV NS1 induces the release of IL-1β in a caspase-1 dependent manner. Additionally, we find that DENV NS1-induced inflammasome activation is independent of the NLRP3, Pyrin, and AIM2 inflammasome pathways, but requires CD14. Intriguingly, DENV NS1-induced inflammasome activation does not induce pyroptosis and rapid cell death; instead, macrophages maintain cellular viability while releasing IL-1β. Lastly, we show that caspase-1/11-deficient, but not NLRP3-deficient, mice are more susceptible to lethal DENV infection. Together, these results indicate that the inflammasome pathway acts as a sensor of DENV NS1 and plays a protective role during infection.
Collapse
Affiliation(s)
- Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Evan Y W Juan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sai S Chelluri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Phoebe Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
29
|
Camarão AAR, Gern OL, Stegmann F, Mulenge F, Costa B, Saremi B, Jung K, Lepenies B, Kalinke U, Steffen I. Secreted NS1 proteins of tick-borne encephalitis virus and West Nile virus block dendritic cell activation and effector functions. Microbiol Spectr 2023; 11:e0219223. [PMID: 37707204 PMCID: PMC10581055 DOI: 10.1128/spectrum.02192-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/13/2023] [Indexed: 09/15/2023] Open
Abstract
The flavivirus non-structural protein 1 (NS1) is secreted from infected cells into the circulation and the serum levels correlate with disease severity. The effect of secreted NS1 (sNS1) on non-infected mammalian immune cells is largely unknown. Here, we expressed recombinant sNS1 proteins of tick-borne encephalitis virus (TBEV) and West Nile virus (WNV) and investigated their effects on dendritic cell (DC) effector functions. Murine bone marrow-derived DCs (BMDCs) showed reduced surface expression of co-stimulatory molecules and decreased release of pro-inflammatory cytokines when treated with sNS1 of TBEV or WNV prior to poly(I:C) stimulation. Transcriptional profiles of BMDCs that were sNS1-exposed prior to poly(I:C) stimulation showed two gene clusters that were downregulated by TBEV or WNV sNS1 and that were associated with innate and adaptive immune responses. Functionally, both sNS1 proteins modulated the capacity for BMDCs to induce specific T-cell responses as indicated by reduced IFN-γ levels in both CD4+ and CD8+ T cells after BMDC co-cultivation. In human monocyte-derived DCs, poly(I:C)-induced upregulation of co-stimulatory molecules and cytokine responses were even more strongly impaired by TBEV sNS1 or WNV sNS1 pretreatment than in the murine system. Our findings indicate that exogenous flaviviral sNS1 proteins interfere with DC-mediated stimulation of T cells, which is crucial for the initiation of cell-mediated adaptive immune responses in human flavivirus infections. Collectively, our data determine soluble flaviviral NS1 as a virulence factor responsible for a dampened immune response to flavivirus infections. IMPORTANCE The effective initiation of protective host immune responses controls the outcome of infection, and dysfunctional T-cell responses have previously been associated with symptomatic human flavivirus infections. We demonstrate that secreted flavivirus NS1 proteins modulate innate immune responses of uninfected bystander cells. In particular, sNS1 markedly reduced the capacity of dendritic cells to stimulate T-cell responses upon activation. Hence, by modulating cellular host responses that are required for effective antigen presentation and initiation of adaptive immunity, sNS1 proteins may contribute to severe outcomes of flavivirus disease.
Collapse
Affiliation(s)
- António A. R. Camarão
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Olivia Luise Gern
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Felix Stegmann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute for Immunology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Bibiana Costa
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Babak Saremi
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Bernd Lepenies
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute for Immunology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
- Cluster of Excellence—Resolving Infection Susceptibility (RESIST, EXC 2155), Hannover Medical School, Hannover, Germany
| | - Imke Steffen
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
30
|
Kapuganti SK, Saumya KU, Verma D, Giri R. Investigating the aggregation perspective of Dengue virus proteome. Virology 2023; 586:12-22. [PMID: 37473502 DOI: 10.1016/j.virol.2023.07.010] [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: 04/24/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023]
Abstract
Dengue viruses are human pathogens that are transmitted through mosquitoes. Apart from the typical symptoms associated with viral fevers, DENV infections are known to cause several neurological complications such as meningitis, encephalitis, intracranial haemorrhage, retinopathies along with the more severe, and sometimes fatal, vascular leakage and dengue shock syndrome. This study was designed to investigate, in detail, the predicted viral protein aggregation prone regions among all serotypes. Further, in order to understand the cross-talk between viral protein aggregation and aggregation of cellular proteins, cross-seeding experiments between the DENV NS1 (1-30), corresponding to the β-roll domain and the diabetes hallmark protein, amylin, were performed. Various techniques such as fluorescence spectroscopy, circular dichroism, atomic force microscopy and immunoblotting have been employed for this. We observe that the DENV proteomes have many predicted APRs and the NS1 (1-30) of DENV1-3, 2K and capsid anchor of DENV2 and DENV4 are capable of forming amyloids, in vitro. Further, the DENV NS1 (1-30), aggregates are also able to cross-seed and enhance amylin aggregation and vice-versa. This knowledge may lead to an opportunity for designing suitable inhibitors of protein aggregation that may be beneficial for viral infections and comorbidities.
Collapse
Affiliation(s)
- Shivani Krishna Kapuganti
- Indian Institute of Technology Mandi, School of Basic Sciences, VPO Kamand, Himachal Pradesh, 175005, India
| | - Kumar Udit Saumya
- Indian Institute of Technology Mandi, School of Basic Sciences, VPO Kamand, Himachal Pradesh, 175005, India
| | - Deepanshu Verma
- Indian Institute of Technology Mandi, School of Basic Sciences, VPO Kamand, Himachal Pradesh, 175005, India
| | - Rajanish Giri
- Indian Institute of Technology Mandi, School of Basic Sciences, VPO Kamand, Himachal Pradesh, 175005, India.
| |
Collapse
|
31
|
Ramu ST, Dissanayake M, Jeewandara C, Bary F, Harvie M, Gomes L, Wijesinghe A, Ariyaratne D, Ogg GS, Malavige GN. Antibody and memory B cell responses to the dengue virus NS1 antigen in individuals with varying severity of past infection. Immunology 2023; 170:47-59. [PMID: 37075785 PMCID: PMC11495261 DOI: 10.1111/imm.13651] [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: 09/02/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
To further understand the role of NS1-specific antibodies (Abs) in disease pathogenesis, we compared neutralizing antibody levels (Nabs), NS1-Ab levels, IgG antibody subclass profiles and NS1-specific memory B-cell responses (Bmems) in individuals, with varying severity of past dengue. Nabs (Neut50 titres) were assessed using the Foci Reduction Neutralization Test (FRNT) and in-house ELISAs were used to assess NS1-Abs and NS1-Ab subclasses for all four DENV serotypes in individuals with past DF (n = 22), those with past DHF (n = 14) and seronegative (SN) individuals (n = 7). B-cell ELISpot assays were used to assess NS1-specific Bmem responses. 15/22 (68.18%) individuals with past DF and 9/14 (64.29%) individuals with past DHF had heterotypic infections. Neut50 titres were found to be significantly higher for DENV1 than DENV2 (p = 0.0006) and DENV4 (p = 0.0127), in those with past DHF, whereas there was no significant difference seen in titres for different DENV serotypes in those with past DF. Overall NS1-Ab to all serotypes and NS1-specific IgG1 responses for DENV1, 2 and 4 serotypes were significantly higher in those with past DHF than individuals with past DF. Those with past DHF also had higher IgG1 than IgG3 for DENV1 and DENV3, whereas no differences were seen in those with past DF. Over 50% of those with past DF or DHF had NS1-specific Bmem responses to >2 DENV serotypes. There was no difference in the frequency of Bmem responses to any of the DENV serotypes between individuals with past DF and DHF. Although the frequency of Bmem responses to DENV1 correlated with DENV1-specific NS1-Abs levels (Spearman r = 0.35, p = 0.02), there was no correlation with other DENV serotypes. We found that those with past DF had broadly cross-reactive Nabs, while those with past DHF had higher NS1-Ab responses possibly with a different functionality profile than those with past DF. Therefore, it would be important to further evaluate the functionality of NS1-specific antibody and Bmem responses to find out the type of antibody repertoire that is associated with protection against severe disease.
Collapse
Affiliation(s)
- Shyrar Tanussiya Ramu
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Madushika Dissanayake
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Farha Bary
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Michael Harvie
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Laksiri Gomes
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Ayesha Wijesinghe
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Dinuka Ariyaratne
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
| | - Graham S. Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Gathsaurie Neelika Malavige
- Allergy Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, Faculty of Medical SciencesUniversity of Sri JayewardenepuraColomboSri Lanka
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
32
|
Alcalá AC, Ludert JE. The dengue virus NS1 protein; new roles in pathogenesis due to similarities with and affinity for the high-density lipoprotein (HDL)? PLoS Pathog 2023; 19:e1011587. [PMID: 37616216 PMCID: PMC10449462 DOI: 10.1371/journal.ppat.1011587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023] Open
Affiliation(s)
- Ana C. Alcalá
- MU Center for Influenza and Emerging Infectious Diseases, University of Missouri, Columbia, MO, United States of America
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States of America
- Bond Life Science Center, University of Missouri, Columbia, MO, United States of America
| | - Juan E. Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
33
|
Zhang S, He Y, Wu Z, Wang M, Jia R, Zhu D, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Gao Q, Sun D, Zhang L, Yu Y, Chen S, Cheng A. Secretory pathways and multiple functions of nonstructural protein 1 in flavivirus infection. Front Immunol 2023; 14:1205002. [PMID: 37520540 PMCID: PMC10372224 DOI: 10.3389/fimmu.2023.1205002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The genus Flavivirus contains a wide variety of viruses that cause severe disease in humans, including dengue virus, yellow fever virus, Zika virus, West Nile virus, Japanese encephalitis virus and tick-borne encephalitis virus. Nonstructural protein 1 (NS1) is a glycoprotein that encodes a 352-amino-acid polypeptide and has a molecular weight of 46-55 kDa depending on its glycosylation status. NS1 is highly conserved among multiple flaviviruses and occurs in distinct forms, including a dimeric form within the endoplasmic reticulum, a cell-associated form on the plasma membrane, or a secreted hexameric form (sNS1) trafficked to the extracellular matrix. Intracellular dimeric NS1 interacts with other NSs to participate in viral replication and virion maturation, while extracellular sNS1 plays a critical role in immune evasion, flavivirus pathogenesis and interactions with natural vectors. In this review, we provide an overview of recent research progress on flavivirus NS1, including research on the structural details, the secretory pathways in mammalian and mosquito cells and the multiple functions in viral replication, immune evasion, pathogenesis and interaction with natural hosts, drawing together the previous data to determine the properties of this protein.
Collapse
Affiliation(s)
- Senzhao Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Yu He
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Zhen Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Qun Gao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Di Sun
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Ling Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yanling Yu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, China
| |
Collapse
|
34
|
Malnero CM, Azevedo RC, Bergmann IE, de Meneses MDF, Cavalcanti AC, Ibáñez LI, Malirat V. Expression of recombinant dengue virus type 1 non-structural protein 1 in mammalian cells and preliminary assessment of its suitability to detect human IgG antibodies elicited by viral infection. J Immunol Methods 2023; 518:113503. [PMID: 37263391 DOI: 10.1016/j.jim.2023.113503] [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: 12/14/2022] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/03/2023]
Abstract
In recent years dengue has become a rapidly growing public health problem worldwide, however, the availability of accurate and affordable diagnostic immunoassays is limited, partly due to the difficulty of producing large quantities of purified antigen. Non-structural protein 1 (NS1) has shown to be a good candidate for inclusion in diagnostic assays and for serosurveys, particularly in endemic countries as a prerequisite for vaccination. In this work the NS1 antigen derived from dengue virus type-1 (DENV1) was expressed in HEK293-T cells and purified by affinity chromatography. The recombinant protein was recovered properly folded as dimers, highly purified and with good yield (1.5 mg/L). It was applied as a serological probe in an indirect ELISA developed in this work to detect human IgG antibodies. Preliminary comparative performance values of 81.1% sensitivity and 83.0% specificity of the developed and preliminary validated iELISA, relative to a commercial kit were obtained, suggesting that the purified recombinant DENV1 NS1 antigen is suitable to detect IgG antibodies, indicative of past DENV infection.
Collapse
Affiliation(s)
- Cristian Miguel Malnero
- Centro de Virología Humana y Animal (CEVHAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Abierta Interamericana (UAI), Buenos Aires C1287, Argentina
| | - Renata Campos Azevedo
- Department of Virology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ingrid Evelyn Bergmann
- Centro de Virología Humana y Animal (CEVHAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Abierta Interamericana (UAI), Buenos Aires C1287, Argentina
| | | | - Andrea Cony Cavalcanti
- Department of Virology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Central Laboratory of Public Health Noel Nutels (LACEN-RJ), Rio de Janeiro, Brazil
| | - Lorena Itatí Ibáñez
- Instituto de Química Física de los Materiales, Medio Ambiente y Energía (INQUIMAE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Buenos Aires C1428EGA, Argentina
| | - Viviana Malirat
- Centro de Virología Humana y Animal (CEVHAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Abierta Interamericana (UAI), Buenos Aires C1287, Argentina.
| |
Collapse
|
35
|
Tan BEK, Beard MR, Eyre NS. Identification of Key Residues in Dengue Virus NS1 Protein That Are Essential for Its Secretion. Viruses 2023; 15:v15051102. [PMID: 37243188 DOI: 10.3390/v15051102] [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: 02/24/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Dengue virus (DENV) non-structural protein 1 (NS1) is involved in multiple aspects of the DENV lifecycle. Importantly, it is secreted from infected cells as a hexameric lipoparticle that mediates vascular damage that is a hallmark of severe dengue. Although the secretion of NS1 is known to be important in DENV pathogenesis, the exact molecular features of NS1 that are required for its secretion from cells are not fully understood. In this study, we employed random point mutagenesis in the context of an NS1 expression vector encoding a C-terminal HiBiT luminescent peptide tag to identify residues within NS1 that are essential for its secretion. Using this approach, we identified 10 point mutations that corresponded with impaired NS1 secretion, with in silico analyses indicating that the majority of these mutations are located within the β-ladder domain. Additional studies on two of these mutants, V220D and A248V, revealed that they prevented viral RNA replication, while studies using a DENV NS1-NS5 viral polyprotein expression system demonstrated that these mutations resulted in a more reticular NS1 localisation pattern and failure to detect mature NS1 at its predicted molecular weight by Western blotting using a conformation-specific monoclonal antibody. Together, these studies demonstrate that the combination of a luminescent peptide tagged NS1 expression system with random point mutagenesis enables rapid identification of mutations that alter NS1 secretion. Two such mutations identified via this approach revealed residues that are essential for correct NS1 processing or maturation and viral RNA replication.
Collapse
Affiliation(s)
- Brandon E K Tan
- Research Centre of Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Michael R Beard
- Research Centre of Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Nicholas S Eyre
- College of Medicine and Public Health (CMPH), Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
36
|
van den Elsen K, Chew BLA, Ho JS, Luo D. Flavivirus nonstructural proteins and replication complexes as antiviral drug targets. Curr Opin Virol 2023; 59:101305. [PMID: 36870091 PMCID: PMC10023477 DOI: 10.1016/j.coviro.2023.101305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/02/2023] [Accepted: 01/17/2023] [Indexed: 03/06/2023]
Abstract
Many flaviviruses are well-known pathogens, such as dengue, Zika, Japanese encephalitis, and yellow fever viruses. Among them, dengue viruses cause global epidemics and threaten billions of people. Effective vaccines and antivirals are in desperate need. In this review, we focus on the recent advances in understanding viral nonstructural (NS) proteins as antiviral drug targets. We briefly summarize the experimental structures and predicted models of flaviviral NS proteins and their functions. We highlight a few well-characterized inhibitors targeting these NS proteins and provide an update about the latest development. NS4B emerges as one of the most promising drug targets as novel inhibitors targeting NS4B and its interaction network are entering clinical studies. Studies aiming to elucidate the architecture and molecular basis of viral replication will offer new opportunities for novel antiviral discovery. Direct-acting agents against dengue and other pathogenic flaviviruses may be available very soon.
Collapse
Affiliation(s)
- Kaïn van den Elsen
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore 636921, Singapore; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Bing Liang Alvin Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore 636921, Singapore; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Jun Sheng Ho
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore 636921, Singapore; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, EMB 03-07, 59 Nanyang Drive, Singapore 636921, Singapore; NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore.
| |
Collapse
|
37
|
Wilken L, Stelz S, Agac A, Sutter G, Prajeeth CK, Rimmelzwaan GF. Recombinant Modified Vaccinia Virus Ankara Expressing a Glycosylation Mutant of Dengue Virus NS1 Induces Specific Antibody and T-Cell Responses in Mice. Vaccines (Basel) 2023; 11:vaccines11040714. [PMID: 37112626 PMCID: PMC10140942 DOI: 10.3390/vaccines11040714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
The four serotypes of dengue virus (DENV1-4) continue to pose a major public health threat. The first licenced dengue vaccine, which expresses the surface proteins of DENV1-4, has performed poorly in immunologically naïve individuals, sensitising them to antibody-enhanced dengue disease. DENV non-structural protein 1 (NS1) can directly induce vascular leakage, the hallmark of severe dengue disease, which is blocked by NS1-specific antibodies, making it an attractive target for vaccine development. However, the intrinsic ability of NS1 to trigger vascular leakage is a potential drawback of its use as a vaccine antigen. Here, we modified DENV2 NS1 by mutating an N-linked glycosylation site associated with NS1-induced endothelial hyperpermeability and used modified vaccinia virus Ankara (MVA) as a vector for its delivery. The resulting construct, rMVA-D2-NS1-N207Q, displayed high genetic stability and drove efficient secretion of NS1-N207Q from infected cells. Secreted NS1-N207Q was composed of dimers and lacked N-linked glycosylation at position 207. Prime-boost immunisation of C57BL/6J mice induced high levels of NS1-specific antibodies binding various conformations of NS1 and elicited NS1-specific CD4+ T-cell responses. Our findings support rMVA-D2-NS1-N207Q as a promising and potentially safer alternative to existing NS1-based vaccine candidates, warranting further pre-clinical testing in a relevant mouse model of DENV infection.
Collapse
Affiliation(s)
- Lucas Wilken
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Sonja Stelz
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Ayse Agac
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig Maximilian University (LMU), 80539 Munich, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| |
Collapse
|
38
|
Sharma A, Krishna S, Sowdhamini R. Bioinformatics Analysis of Mutations Sheds Light on the Evolution of Dengue NS1 Protein With Implications in the Identification of Potential Functional and Druggable Sites. Mol Biol Evol 2023; 40:7043264. [PMID: 36795614 PMCID: PMC9989740 DOI: 10.1093/molbev/msad033] [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: 08/30/2022] [Revised: 12/26/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
Non-structural protein (NS1) is a 350 amino acid long conserved protein in the dengue virus. Conservation of NS1 is expected due to its importance in dengue pathogenesis. The protein is known to exist in dimeric and hexameric states. The dimeric state is involved in its interaction with host proteins and viral replication, and the hexameric state is involved in viral invasion. In this work, we performed extensive structure and sequence analysis of NS1 protein, and uncovered the role of NS1 quaternary states in its evolution. A three-dimensional modeling of unresolved loop regions in NS1 structure is performed. "Conserved" and "Variable" regions within NS1 protein were identified from sequences obtained from patient samples and the role of compensatory mutations in selecting destabilizing mutations were identified. Molecular dynamics (MD) simulations were performed to extensively study the effect of a few mutations on NS1 structure stability and compensatory mutations. Virtual saturation mutagenesis, predicting the effect of every individual amino acid substitution on NS1 stability sequentially, revealed virtual-conserved and variable sites. The increase in number of observed and virtual-conserved regions across NS1 quaternary states suggest the role of higher order structure formation in its evolutionary conservation. Our sequence and structure analysis could enable in identifying possible protein-protein interfaces and druggable sites. Virtual screening of nearly 10,000 small molecules, including FDA-approved drugs, permitted us to recognize six drug-like molecules targeting the dimeric sites. These molecules could be promising due to their stable interactions with NS1 throughout the simulation.
Collapse
Affiliation(s)
- Abhishek Sharma
- National Centre for Biological Science, TIFR, Bangalore, India
| | - Sudhir Krishna
- National Centre for Biological Science, TIFR, Bangalore, India.,Department of School of Interdisciplinary Life Sciences, Indian Institute of Technology Goa, Farmagudi, Pond-403401, Goa, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Science, TIFR, Bangalore, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.,Computational Biology, Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| |
Collapse
|
39
|
Prudencio CR, Gomes da Costa V, Rocha LB, da Costa HHM, Orts DJB, da Silva Santos FR, Rahal P, Lino NAB, da Conceição PJP, Bittar C, Machado RRG, Durigon EL, Araujo JP, Polatto JM, da Silva MA, de Oliveira JA, Mitsunari T, Pereira LR, Andreata-Santos R, de Souza Ferreira LC, Luz D, Piazza RMF. Identification of Zika Virus NS1-Derived Peptides with Potential Applications in Serological Tests. Viruses 2023; 15:v15030654. [PMID: 36992364 PMCID: PMC10052002 DOI: 10.3390/v15030654] [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: 12/21/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne pathogen, is an emerging arbovirus associated with sporadic symptomatic cases of great medical concern, particularly among pregnant women and newborns affected with neurological disorders. Serological diagnosis of ZIKV infection is still an unmet challenge due to the co-circulation of the dengue virus, which shares extensive sequence conservation of structural proteins leading to the generation of cross-reactive antibodies. In this study, we aimed to obtain tools for the development of improved serological tests for the detection of ZIKV infection. Polyclonal sera (pAb) and a monoclonal antibody (mAb 2F2) against a recombinant form of the ZIKV nonstructural protein 1 (NS1) allowed the identification of linear peptide epitopes of the NS1 protein. Based on these findings, six chemically synthesized peptides were tested both in dot blot and ELISA assays using convalescent sera collected from ZIKV-infected patients. Two of these peptides specifically detected the presence of ZIKV antibodies and proved to be candidates for the detection of ZIKV-infected subjects. The availability of these tools opens perspectives for the development of NS1-based serological tests with enhanced sensitivity regarding other flaviviruses.
Collapse
Affiliation(s)
- Carlos Roberto Prudencio
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Vivaldo Gomes da Costa
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Leticia Barboza Rocha
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Hernan Hermes Monteiro da Costa
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Diego José Belato Orts
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Felipe Rocha da Silva Santos
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Paula Rahal
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Nikolas Alexander Borsato Lino
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Pâmela Jóyce Previdelli da Conceição
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Cintia Bittar
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Rafael Rahal Guaragna Machado
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Edison Luiz Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - João Pessoa Araujo
- Instituto de Biotecnologia, Universidade Estadual Paulista Júlio de Mesquita Filho, Botucatu 18607-440, SP, Brazil
| | - Juliana Moutinho Polatto
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Miriam Aparecida da Silva
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Joyce Araújo de Oliveira
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Thais Mitsunari
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Lennon Ramos Pereira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Robert Andreata-Santos
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Luís Carlos de Souza Ferreira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
- Plataforma Científica Pasteur USP, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Daniela Luz
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Roxane Maria Fontes Piazza
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| |
Collapse
|
40
|
Dolata KM, Fuchs W, Caignard G, Dupré J, Pannhorst K, Blome S, Mettenleiter TC, Karger A. CP204L Is a Multifunctional Protein of African Swine Fever Virus That Interacts with the VPS39 Subunit of the Homotypic Fusion and Vacuole Protein Sorting Complex and Promotes Lysosome Clustering. J Virol 2023; 97:e0194322. [PMID: 36722971 PMCID: PMC9972913 DOI: 10.1128/jvi.01943-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 01/06/2023] [Indexed: 02/02/2023] Open
Abstract
Virus replication depends on a complex interplay between viral and host proteins. In the case of African swine fever virus (ASFV), a large DNA virus, only a few virus-host protein-protein interactions have been identified to date. In this study, we demonstrate that the ASFV protein CP204L interacts with the cellular homotypic fusion and protein sorting (HOPS) protein VPS39, blocking its association with the lysosomal HOPS complex, which modulates endolysosomal trafficking and promotes lysosome clustering. Instead, CP204L and VPS39 are targeted to virus factories and localized at the periphery of the virus DNA replication sites. Furthermore, we show that loss of VPS39 reduces the levels of virus proteins synthesized in the early phase of infection and delays ASFV replication but does not completely inhibit it. Collectively, these results identify a novel virus-host protein interaction that modulates host membrane rearrangement during infection and provide evidence that CP204L is a multifunctional protein engaged in distinct steps of the ASFV life cycle. IMPORTANCE African swine fever virus (ASFV) was first identified over a hundred years ago. Since then, much effort has been made to understand the pathogenesis of ASFV. However, the specific roles of many individual ASFV proteins during the infection remain enigmatic. This study provides evidence that CP204L, one of the most abundant ASFV proteins, modulates endosomal trafficking during virus infection. Through protein-protein interaction, CP204L prevents the recruitment of VPS39 to the endosomal and lysosomal membranes, resulting in their accumulation. Consequently, CP204L and VPS39 become sequestered in the ASFV replication and assembly site, known as the virus factory. These results uncover a novel function of viral protein CP204L and extend our understanding of complex interaction between virus and host.
Collapse
Affiliation(s)
- Katarzyna Magdalena Dolata
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Walter Fuchs
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Grégory Caignard
- UMR Virologie, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale d’Alfort, Anses, Université Paris-Est, Maisons-Alfort, France
| | - Juliette Dupré
- UMR Virologie, INRAE, Ecole Nationale Vétérinaire d’Alfort, Laboratoire de Santé Animale d’Alfort, Anses, Université Paris-Est, Maisons-Alfort, France
| | - Katrin Pannhorst
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Sandra Blome
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Thomas C. Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Axel Karger
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| |
Collapse
|
41
|
Poveda Cuevas SA, Barroso da Silva FL, Etchebest C. NS1 from Two Zika Virus Strains Differently Interact with a Membrane: Insights to Understand Their Differential Virulence. J Chem Inf Model 2023; 63:1386-1400. [PMID: 36780300 DOI: 10.1021/acs.jcim.2c01461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Zika virus (ZIKV) from Uganda (UG) expresses a phenotype related to fetal loss, whereas the variant from Brazil (BR) induces microcephaly in neonates. The differential virulence has a direct relation to biomolecular mechanisms that make one strain more aggressive than the other. The nonstructural protein 1 (NS1) is a key viral toxin to comprehend these viral discrepancies because of its versatility in many processes of the virus life cycle. Here, we aim to examine through coarse-grained models and molecular dynamics simulations the protein-membrane interactions for both NS1ZIKV-UG and NS1ZIKV-BR dimers. A first evaluation allowed us to establish that the NS1 proteins, in the membrane presence, explore new conformational spaces when compared to systems simulated without a lipid bilayer. These events derive from both differential coupling patterns and discrepant binding affinities to the membrane. The N-terminal domain, intertwined loop, and greasy finger proposed previously as binding membrane regions were also computationally confirmed by us. The anchoring sites have aromatic and ionizable residues that manage the assembly of NS1 toward the membrane, especially for the Ugandan variant. Furthermore, in the presence of the membrane, the difference in the dynamic cross-correlation of residues between the two strains is particularly pronounced in the putative epitope regions. This suggests that the protein-membrane interaction induces changes in the distal part related to putative epitopes. Taken together, these results open up new strategies for the treatment of flaviviruses that would specifically target these dynamic differences.
Collapse
Affiliation(s)
- Sergio Alejandro Poveda Cuevas
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, Frankfurt am Main, Hesse DE-60590, Germany.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Fernando L Barroso da Silva
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Catherine Etchebest
- Université Paris Cité and Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, F-75015 Paris, France.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| |
Collapse
|
42
|
Ogire E, El-Kalamouni C, Desprès P, Roche M. Stability of Dengue 2 Nonstructural Glycoprotein 1 (NS1) Is Affected by the Nature of Basic Residue at Position NS1-324. Curr Issues Mol Biol 2023; 45:1644-1654. [PMID: 36826051 PMCID: PMC9955058 DOI: 10.3390/cimb45020106] [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] [Received: 12/03/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 02/17/2023] Open
Abstract
Dengue is the most prevalent mosquito-borne viral disease. It is caused by the infection of any of the four dengue virus (DENV) serotypes DENV-1 to DENV-4. The DENV non-structural glycoprotein 1 (NS1) plays an important role in virus replication and the immunopathogenesis of virus infection. The NS1 protein has been identified as both a cell-associated homodimer and a soluble secreted lipoprotein nanoparticle. The nature of the residues at positions NS1-272 and NS1-324 in the β-ladder domain may have an effect on the biological behaviors of DENV-2 NS1 protein in human hepatoma Huh7 cells. The stability of the NS1 protein from the Reunion 2018 DENV-2 strain was affected by the presence of lysine residues at positions 272 and 324. In the present study, we evaluated the impact of mutations into lysine at positions 272 and 324 on recombinant NS1 protein from the DES-14 DENV-2 strain bearing arginine residue on these two positions. The DES-14 NS1 protein mutant bearing a lysine at position 324 was deficient in protein stability and secretion compared to wild-type protein. The defect in the DES-14 NS1 protein mutant was associated to oxidative stress and pro-inflammatory cytokine activation in Huh7 cells. The ubiquitin-proteasome proteolytic pathway might play a key role in the stability of DENV-2 protein bearing a lysine residue at position 324.
Collapse
Affiliation(s)
| | | | | | - Marjolaine Roche
- Unité Mixte de Recherche Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de La Réunion, INSERM U1187, CNRS 9192, IRD 249, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France
| |
Collapse
|
43
|
Activation of Early Proinflammatory Responses by TBEV NS1 Varies between the Strains of Various Subtypes. Int J Mol Sci 2023; 24:ijms24021011. [PMID: 36674524 PMCID: PMC9863113 DOI: 10.3390/ijms24021011] [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] [Received: 12/09/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Tick-borne encephalitis (TBE) is an emerging zoonosis that may cause long-term neurological sequelae or even death. Thus, there is a growing interest in understanding the factors of TBE pathogenesis. Viral genetic determinants may greatly affect the severity and consequences of TBE. In this study, nonstructural protein 1 (NS1) of the tick-borne encephalitis virus (TBEV) was tested as such a determinant. NS1s of three strains with similar neuroinvasiveness belonging to the European, Siberian and Far-Eastern subtypes of TBEV were studied. Transfection of mouse cells with plasmids encoding NS1 of the three TBEV subtypes led to different levels of NS1 protein accumulation in and secretion from the cells. NS1s of TBEV were able to trigger cytokine production either in isolated mouse splenocytes or in mice after delivery of NS1 encoding plasmids. The profile and dynamics of TNF-α, IL-6, IL-10 and IFN-γ differed between the strains. These results demonstrated the involvement of TBEV NS1 in triggering an immune response and indicated the diversity of NS1 as one of the genetic factors of TBEV pathogenicity.
Collapse
|
44
|
Shu B, Ooi JSG, Tan AWK, Ng TS, Dejnirattisai W, Mongkolsapaya J, Fibriansah G, Shi J, Kostyuchenko VA, Screaton GR, Lok SM. CryoEM structures of the multimeric secreted NS1, a major factor for dengue hemorrhagic fever. Nat Commun 2022; 13:6756. [PMID: 36347841 PMCID: PMC9643530 DOI: 10.1038/s41467-022-34415-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 10/25/2022] [Indexed: 11/11/2022] Open
Abstract
Dengue virus infection can cause dengue hemorrhagic fever (DHF). Dengue NS1 is multifunctional. The intracellular dimeric NS1 (iNS1) forms part of the viral replication complex. Previous studies suggest the extracellular secreted NS1 (sNS1), which is a major factor contributing to DHF, exists as hexamers. The structure of the iNS1 is well-characterised but not that of sNS1. Here we show by cryoEM that the recombinant sNS1 exists in multiple oligomeric states: the tetrameric (stable and loose conformation) and hexameric structures. Stability of the stable and loose tetramers is determined by the conformation of their N-terminal domain - elongated β-sheet or β-roll. Binding of an anti-NS1 Fab breaks the loose tetrameric and hexameric sNS1 into dimers, whereas the stable tetramer remains largely unbound. Our results show detailed quaternary organization of different oligomeric states of sNS1 and will contribute towards the design of dengue therapeutics.
Collapse
Affiliation(s)
- Bo Shu
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Justin S G Ooi
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Aaron W K Tan
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Thiam-Seng Ng
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | | | | | - Guntur Fibriansah
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Jian Shi
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Victor A Kostyuchenko
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore
| | - Gavin R Screaton
- Medical Sciences Division, University of Oxford, Oxford, OX3 9D, UK
| | - Shee-Mei Lok
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore.
- Centre for Bioimaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, 117557, Singapore.
| |
Collapse
|
45
|
Molecular Determinants of Tissue Specificity of Flavivirus Nonstructural Protein 1 Interaction with Endothelial Cells. J Virol 2022; 96:e0066122. [PMID: 36106873 PMCID: PMC9555157 DOI: 10.1128/jvi.00661-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Members of the mosquito-borne flavivirus genus such as dengue (DENV), West Nile (WNV), and Zika (ZIKV) viruses cause distinct diseases and affect different tissues. We previously found that the secreted flaviviral nonstructural protein 1 (NS1) interacts with endothelial cells and disrupts endothelial barrier function in a tissue-specific manner consistent with the disease tropism of the respective viruses. However, the underlying molecular mechanism of this tissue-specific NS1-endothelial cell interaction is not well understood. To elucidate the distinct role(s) that the wing and β-ladder domains of NS1 play in NS1 interactions with endothelial cells, we constructed flavivirus NS1 chimeras that exchanged the wing and β-ladder domains in a pairwise manner between DENV, WNV, and ZIKV NS1. We found that both the NS1 wing and β-ladder domains conferred NS1 tissue-specific endothelial dysfunction, with the wing conferring cell binding and the β-ladder involved in inducing endothelial hyperpermeability as measured by transendothelial electrical resistance. To narrow down the amino acids dictating cell binding specificity, we utilized the DENV-WNV NS1 chimera and identified residues 91 to 93 (GDI) of DENV NS1 as a molecular motif determining binding specificity. Further, using an in vivo mouse model of localized leak, we found that the GDI motif of the wing domain was essential for triggering DENV NS1-induced vascular leak in mouse dermis. Taken together, we identify molecular determinants of flavivirus NS1 that confer NS1 binding and vascular leak and highlight the importance of the NS1 wing domain for flavivirus pathogenesis. IMPORTANCE Flavivirus NS1 is secreted into the bloodstream from infected cells during a viral infection. Dengue virus NS1 contributes to severe dengue pathology such as endothelial dysfunction and vascular leak independently of the virus. We have shown that multiple flavivirus NS1 proteins result in endothelial dysfunction in a tissue-specific manner consistent with their respective viral tropism. Here, we aimed to identify the molecular determinants that make some, but not other, flavivirus NS1 proteins bind to select endothelial cells in vitro and cause vascular leak in a mouse model. We identified the wing domain of NS1 as a primary determinant conferring differential endothelial dysfunction and vascular leak and narrowed the contributing amino acid residues to a three-residue motif within the wing domain. The insights from this study pave the way for future studies on the effects of flavivirus NS1 on viral dissemination and pathogenesis and offer potential new avenues for antiviral therapies.
Collapse
|
46
|
Almeida NBF, Sousa TASL, Santos VCF, Lacerda CMS, Silva TG, Grenfell RFQ, Plentz F, Andrade ASR. DNA aptamer selection and construction of an aptasensor based on graphene FETs for Zika virus NS1 protein detection. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2022; 13:873-881. [PMID: 36105684 PMCID: PMC9443353 DOI: 10.3762/bjnano.13.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that is phylogenetically close to other medically important flaviviruses with high global public health significance, such as dengue (DENV) and yellow fever (YFV) viruses. Correct diagnosis of a flavivirus infection can be challenging, particularly in world regions where more than one flavivirus co-circulates and YFV vaccination is mandatory. Acid nucleic aptamers are oligonucleotides that bind to a specific target molecule with high affinity and specificity. Because of their unique characteristics, aptamers are promising tools for biosensor development. Aptamers are usually obtained through a procedure called "systematic evolution of ligands by exponential enrichment" (SELEX). In this study, we select an aptamer (termed ZIKV60) by capillary electrophoresis SELEX (CE-SELEX) to the Zika virus non-structural protein 1 (NS1) and counterselection against the NS1 proteins of DENV (serotypes 1, 2, 3, and 4) and YFV. The ZIKV60 dissociation constant (K d) is determined by enzyme-linked oligonucleotide assay (ELONA) and the aptamer specificity is evaluated by quantitative real-time polymerase chain reaction. ZIKV60 shows a high binding affinity to the ZIKV NS1 protein with a K d value of 2.28 ± 0.28 nM. The aptamer presents high specificity for ZIKV NS1 compared to NS1 of DENV and YFV. Furthermore, graphene field-effect transistor devices functionalized with ZIKV60 exhibit an evident identification of NS1 protein diluted in human serum. These results point to the applicability of biosensors based on the ZIKV60 aptamer for the differential diagnosis of the Zika virus.
Collapse
Affiliation(s)
- Nathalie B F Almeida
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Thiago A S L Sousa
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
- Current address: DTU Physics, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Viviane C F Santos
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Camila M S Lacerda
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Thais G Silva
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Rafaella F Q Grenfell
- Instituto René Rachou - Fundação Oswaldo Cruz, Avenida Augusto de Lima 1715, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Flavio Plentz
- Departamento de Física, ICEx, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
- MedicOnChip, Parque Tecnológico de Belo Horizonte-BH-TEC, Rua Professor José Vieira de Mendonça 770, Belo Horizonte, CEP 31310-260, Brazil
| | - Antero S R Andrade
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Avenida Presidente Antônio Carlos 6627, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
47
|
Besson B, Lezcano OM, Overheul GJ, Janssen K, Spruijt CG, Vermeulen M, Qu J, van Rij RP. Arbovirus-vector protein interactomics identifies Loquacious as a co-factor for dengue virus replication in Aedes mosquitoes. PLoS Pathog 2022; 18:e1010329. [PMID: 36074777 PMCID: PMC9488832 DOI: 10.1371/journal.ppat.1010329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 09/20/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022] Open
Abstract
Efficient virus replication in Aedes vector mosquitoes is essential for the transmission of arboviral diseases such as dengue virus (DENV) in human populations. Like in vertebrates, virus-host protein-protein interactions are essential for viral replication and immune evasion in the mosquito vector. Here, 79 mosquito host proteins interacting with DENV non-structural proteins NS1 and NS5 were identified by label-free mass spectrometry, followed by a functional screening. We confirmed interactions with host factors previously observed in mammals, such as the oligosaccharyltransferase complex, and we identified protein-protein interactions that seem to be specific for mosquitoes. Among the interactors, the double-stranded RNA (dsRNA) binding protein Loquacious (Loqs), an RNA interference (RNAi) cofactor, was found to be essential for efficient replication of DENV and Zika virus (ZIKV) in mosquito cells. Loqs did not affect viral RNA stability or translation of a DENV replicon and its proviral activity was independent of its RNAi regulatory activity. Interestingly, Loqs colocalized with DENV dsRNA replication intermediates in infected cells and directly interacted with high affinity with DENV RNA in the 3' untranslated region in vitro (KD = 48-62 nM). Our study provides an interactome for DENV NS1 and NS5 and identifies Loqs as a key proviral host factor in mosquitoes. We propose that DENV hijacks a factor of the RNAi mechanism for replication of its own RNA.
Collapse
Affiliation(s)
- Benoit Besson
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Oscar M. Lezcano
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gijs J. Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kirsten Janssen
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cornelia G. Spruijt
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Jieqiong Qu
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald P. van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Benfrid S, Park K, Dellarole M, Voss JE, Tamietti C, Pehau‐Arnaudet G, Raynal B, Brûlé S, England P, Zhang X, Mikhailova A, Hasan M, Ungeheuer M, Petres S, Biering SB, Harris E, Sakuntabhai A, Buchy P, Duong V, Dussart P, Coulibaly F, Bontems F, Rey FA, Flamand M. Dengue virus NS1 protein conveys pro-inflammatory signals by docking onto high-density lipoproteins. EMBO Rep 2022; 23:e53600. [PMID: 35607830 PMCID: PMC10549233 DOI: 10.15252/embr.202153600] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 10/05/2023] Open
Abstract
The dengue virus nonstructural protein 1 (NS1) is a secreted virulence factor that modulates complement, activates immune cells and alters endothelial barriers. The molecular basis of these events remains incompletely understood. Here we describe a functional high affinity complex formed between NS1 and human high-density lipoproteins (HDL). Collapse of the soluble NS1 hexamer upon binding to the lipoprotein particle leads to the anchoring of amphipathic NS1 dimeric subunits into the HDL outer layer. The stable complex can be visualized by electron microscopy as a spherical HDL with rod-shaped NS1 dimers protruding from the surface. We further show that the assembly of NS1-HDL complexes triggers the production of pro-inflammatory cytokines in human primary macrophages while NS1 or HDL alone do not. Finally, we detect NS1 in complex with HDL and low-density lipoprotein (LDL) particles in the plasma of hospitalized dengue patients and observe NS1-apolipoprotein E-positive complexes accumulating overtime. The functional reprogramming of endogenous lipoprotein particles by NS1 as a means to exacerbate systemic inflammation during viral infection provides a new paradigm in dengue pathogenesis.
Collapse
Affiliation(s)
- Souheyla Benfrid
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Université Paris Descartes SorbonneParis CitéFrance
- Present address:
Laboratoire de Santé AnimaleANSES, INRA, ENVA, UMR 1161Université Paris‐EstMaisons‐AlfortFrance
| | - Kyu‐Ho Park
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Present address:
Applied Molecular VirologyInstitut Pasteur KoreaSeongnam‐siKorea
| | - Mariano Dellarole
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Present address:
Virus Biophysics LaboratoryBionanosciences Research Center (CIBION)National Scientific and Technical Research Council (CONICET)Ciudad Autónoma de Buenos AiresArgentina
| | - James E Voss
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Present address:
Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCAUSA
| | - Carole Tamietti
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
| | | | - Bertrand Raynal
- Molecular Biophysics FacilityCNRS UMR 3528Institut PasteurParisFrance
| | - Sébastien Brûlé
- Molecular Biophysics FacilityCNRS UMR 3528Institut PasteurParisFrance
| | - Patrick England
- Molecular Biophysics FacilityCNRS UMR 3528Institut PasteurParisFrance
| | - Xiaokang Zhang
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Present address:
Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulationthe Brain Cognition and Brain Disease Institute (BCBDI)Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen‐Hong Kong Institute of Brain Science‐Shenzhen Fundamental Research InstitutionsShenzhenChina
| | - Anastassia Mikhailova
- HIV Inflammation et PersistanceInstitut PasteurParisFrance
- Present address:
Division of Molecular NeurobiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and ServiceCB UTechSParisFrance
| | | | - Stéphane Petres
- Production and Purification of Recombinant Proteins FacilityInstitut PasteurParisFrance
| | - Scott B Biering
- Division of Infectious Diseases and VaccinologySchool of Public HealthUniversity of CaliforniaBerkeleyCAUSA
| | - Eva Harris
- Division of Infectious Diseases and VaccinologySchool of Public HealthUniversity of CaliforniaBerkeleyCAUSA
| | | | - Philippe Buchy
- Virology UnitInstitut Pasteur du CambodgeInstitut Pasteur International NetworkPhnom PenhCambodia
- Present address:
GlaxoSmithKline Vaccines R&DSingaporeSingapore
| | - Veasna Duong
- Virology UnitInstitut Pasteur du CambodgeInstitut Pasteur International NetworkPhnom PenhCambodia
| | - Philippe Dussart
- Virology UnitInstitut Pasteur du CambodgeInstitut Pasteur International NetworkPhnom PenhCambodia
| | - Fasséli Coulibaly
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
| | - François Bontems
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
- Département de Biologie et Chimie StructuralesInstitut de Chimie des Substances Naturelles, CNRS UPR2301Gif‐sur‐YvetteFrance
| | - Félix A Rey
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
| | - Marie Flamand
- Unité de Virologie StructuraleInstitut Pasteur and CNRS UMR3569ParisFrance
| |
Collapse
|
49
|
Japanese Encephalitis Virus (JEV) NS1' Enhances the Viral Infection of Dendritic Cells (DCs) and Macrophages in Pig Tonsils. Microbiol Spectr 2022; 10:e0114722. [PMID: 35730942 PMCID: PMC9430915 DOI: 10.1128/spectrum.01147-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Pigs are the amplifying hosts of Japanese encephalitis virus (JEV). Currently, the safe and effective live attenuated vaccine made of JEV strain SA14-14-2, which does not express NS1', is widely used in humans and domestic animals to prevent JEV infection. In this study, we constructed the NS1' expression recombinant virus (rA66G) through a single nucleotide mutation in NS2A of JEV strain SA14-14-2. Animal experiments showed that NS1' significantly enhanced JEV infection in pig central nervous system (CNS) and tonsil tissues. Pigs shed virus in oronasal secretions in the JEV rA66G virus inoculation group, indicating that NS1' may facilitate the horizontal transmission of JEV. Additionally, dendritic cells (DCs) and macrophages are the main target cells of JEV infection in pig tonsils, which are an important site of persistent JEV infection. The reduction of major histocompatibility complex class II (MHC II) and activation of inducible nitric oxide synthase (iNOS) in pig tonsils caused by viral infection may create a beneficial environment for persistent JEV infection. These results are of significance for JEV infection in pigs and lay the foundation for future studies of JEV persistent infection in pig tonsils. IMPORTANCE Pigs are amplification hosts for Japanese encephalitis virus (JEV). JEV can persist in the tonsils for months despite the presence of neutralizing antibodies. The present study shows that NS1' increases JEV infection in pig tonsils. In addition, DCs and macrophages in the tonsils are the target cells for JEV infection, and JEV NS1' promotes virus infection in DCs and macrophages. This study reveals a novel function of JEV NS1' protein and lays the foundation for future studies of JEV persistent infection in pig tonsils.
Collapse
|
50
|
Structural dynamics of Zika virus NS1 via a reductionist approach reveal the disordered nature of its β-roll domain in isolation. Virology 2022; 573:72-83. [PMID: 35724498 DOI: 10.1016/j.virol.2022.06.005] [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: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
Abstract
Flavivirus Non-structural 1 (NS1) protein performs multiple functions and it is highly plausible that significant structural and folding dynamics of NS1 might play a role in its multifunctionality. It is important to understand the structural conformations of NS1 and its domains in isolation, possibly highlighting the implications on the overall NS1 protein dynamics. Therefore, we have employed extensively long molecular dynamic (MD) simulations in understanding the dynamics of the three structural domains (i.e., β-roll, wing, and β-ladder) in isolation, as a reductionist approach. We also found that the β-ladder domain is highly flexible, while the β-roll domain is disordered during long simulations. Further, we experimentally validated our findings using CD spectroscopy and confirmed the intrinsically disordered behavior of NS1 β-roll in isolation and lipid mimetic environments. Therefore, we believe this study may have implications for significant dynamics played by NS1 protein, specifically during oligomerization of NS1.
Collapse
|