1
|
Imam MA, Alandijany TA, Felemban HR, Attar RM, Faizo AA, Gattan HS, Dwivedi VD, Azhar EI. Machine learning, network pharmacology, and molecular dynamics reveal potent cyclopeptide inhibitors against dengue virus proteins. Mol Divers 2024:10.1007/s11030-024-10975-w. [PMID: 39227512 DOI: 10.1007/s11030-024-10975-w] [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: 07/12/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
The dengue virus is a major global health hazard responsible for an estimated 390 million diseases yearly. This study focused on identifying cyclopeptide inhibitors for envelope structural proteins E, NS1, NS3, and NS5. Additionally, 5579 cyclopeptides were individually screened against the four target proteins using a machine learning-based quantitative structure-activity relationship model. Subsequently, the best 10 cyclopeptides from each protein were selected for molecular docking with their corresponding proteins. Moreover, the protein-peptide complexes with the highest affinity were subjected to a 100-ns molecular dynamics simulation. The protein-protein complexes exhibited superior structural stability and binding interactions. Based on the results of the MD simulation analyses, which included checking values for Root Mean Square Deviation, Root Mean Square Fluctuation, Principal Component Analysis (PCA), free energy landscapes, and energetic components, it was found that NS5-CP03714 complex is more stable and has stronger binding interactions than NS3-CP02054. PCA and free energy landscape plots have confirmed the higher conformational stability of NS5-CP03714. Analysis of the energetic components revealed that NS5-CP03714 (total binding energy = - 47.19 kcal/mol) exhibits more favorable interaction energies and overall binding energy compared to NS3-CP02054 (total binding energy = - 27.36 kcal/mol), suggesting a stronger and more stable formation of the complex. In addition, the drug-target network of two specific peptides (CP02950 and CP05582) and their associated target proteins were analyzed. This analysis revealed valuable information about their ability to target several proteins and their potential for broad-spectrum activity. Additional experimental investigations are necessary to validate these computational results and assess the efficacy of identified peptide inhibitors in biological systems.
Collapse
Affiliation(s)
- Mohammed A Imam
- Department of Medical Microbiology and Parasitology, Qunfudah Faculty of Medicine, Umm Al-Qura University, Al-Qunfudah, 21961, Saudi Arabia
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Thamir A Alandijany
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Hashim R Felemban
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Roba M Attar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Biological Sciences/Microbiology, Faculty of Science, University of Jeddah, , 21959, Jeddah, Saudi Arabia
| | - Arwa A Faizo
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Hattan S Gattan
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia
| | - Vivek Dhar Dwivedi
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India.
- Bioinformatics Research Division, Quanta Calculus, Greater Noida, India.
| | - Esam I Azhar
- Special Infectious Agents Unit-BSL3, King Fahd Medical Research Center, King Abdulaziz University, 21362, Jeddah, Saudi Arabia.
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21362, Jeddah, Saudi Arabia.
| |
Collapse
|
2
|
Rani NV, Baig MS, Pathak B, Kapoor N, Krishnan A. Mutation of conserved histidine residues of dengue virus envelope protein impairs viral like particle maturation and secretion. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119682. [PMID: 38301907 DOI: 10.1016/j.bbamcr.2024.119682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Dengue virus (DENV) envelope protein plays crucial role in virus entry and maturation of virus during infection. Maturation of DENV occurs in the trans Golgi network at slightly acidic pH which is close to pKa of histidine. When exposed to the acidic environment of the late secretory pathway, dengue virus particles go through a significant conformational change, whereby interactions of structural proteins envelope (E) and prM proteins are reorganised and enable furin protease to cleave prM resulting in mature virus. In order to study the role of histidine of E protein in DENV maturation, we mutated 7 conserved histidine residues of envelope protein and assessed the percent of budding using viral like particle (VLP) system. Histidine mutants; H144A, H244A, H261A and H282A severely disrupted VLP formation without any significant change in expression in cell and its oligomerization ability. Treatment with acidotropic amine reversed the defect for all 4 mutants suggesting that these histidines could be involved in maturation and release. Over expression of capsid protein slightly enhanced VLP release of H244A and H261A. Similarly, furin over expression increased VLP release of these mutants. Co-immunoprecipitation studies revealed that prM and E interaction is lost for H244A, H261A and H282A mutants at acidic pH but not at neutral pH indicating that they could be involved in histidine switch during maturation at acidic pH. Detailed analysis of the mutants could provide novel insights on the interplay of envelop protein during maturation and aid in target for drug development.
Collapse
Affiliation(s)
- N Veena Rani
- School of Sciences, IGNOU, New Delhi 110068, India
| | - Mirza Sarwar Baig
- Centre for Virology, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Bharti Pathak
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India
| | - Neera Kapoor
- School of Sciences, IGNOU, New Delhi 110068, India
| | - Anuja Krishnan
- Department of Molecular Medicine, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
3
|
Maloney BE, Carpio KL, Bilyeu AN, Saunders DRD, Park SL, Pohl AE, Ball NC, Raetz JL, Huang CY, Higgs S, Barrett ADT, Roman-Sosa G, Kenney JL, Vanlandingham DL, Huang YJS. Identification of the flavivirus conserved residues in the envelope protein hinge region for the rational design of a candidate West Nile live-attenuated vaccine. NPJ Vaccines 2023; 8:172. [PMID: 37932282 PMCID: PMC10628263 DOI: 10.1038/s41541-023-00765-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
The flavivirus envelope protein is a class II fusion protein that drives flavivirus-cell membrane fusion. The membrane fusion process is triggered by the conformational change of the E protein from dimer in the virion to trimer, which involves the rearrangement of three domains, EDI, EDII, and EDIII. The movement between EDI and EDII initiates the formation of the E protein trimer. The EDI-EDII hinge region utilizes four motifs to exert the hinge effect at the interdomain region and is crucial for the membrane fusion activity of the E protein. Using West Nile virus (WNV) NY99 strain derived from an infectious clone, we investigated the role of eight flavivirus-conserved hydrophobic residues in the EDI-EDII hinge region in the conformational change of E protein from dimer to trimer and viral entry. Single mutations of the E-A54, E-I130, E-I135, E-I196, and E-Y201 residues affected infectivity. Importantly, the E-A54I and E-Y201P mutations fully attenuated the mouse neuroinvasive phenotype of WNV. The results suggest that multiple flavivirus-conserved hydrophobic residues in the EDI-EDII hinge region play a critical role in the structure-function of the E protein and some contribute to the virulence phenotype of flaviviruses as demonstrated by the attenuation of the mouse neuroinvasive phenotype of WNV. Thus, as a proof of concept, residues in the EDI-EDII hinge region are proposed targets to engineer attenuating mutations for inclusion in the rational design of candidate live-attenuated flavivirus vaccines.
Collapse
Affiliation(s)
- Bailey E Maloney
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Kassandra L Carpio
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Ashley N Bilyeu
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Danielle R D Saunders
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
- Department of Biology, Dean of Faculty, United States Air Force Academy, Colorado Springs, CO, 80840, USA
| | - So Lee Park
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Adrienne E Pohl
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Natalia Costa Ball
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Janae L Raetz
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Claire Y Huang
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Stephen Higgs
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Gleyder Roman-Sosa
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Institute of Virology, University of Veterinary Medicine Hanover, Foundation, Buentewg 17, 30559, Hanover, Germany
| | - Joanie L Kenney
- Division of Vector-borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Dana L Vanlandingham
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA
| | - Yan-Jang S Huang
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
- Biosecurity Research Institute, Kansas State University, Manhattan, KS, 66506, USA.
- Department of Microbiology and Immunology and SUNY Center for Vector-Borne Diseases, Institute of Global Health and Translation Science, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
4
|
Meng JX, Hu QM, Zhang LM, Li N, He YW, Yang ZX, Sun Y, Wang JL. Isolation and Genetic Evolution of Dengue Virus from the 2019 Outbreak in Xishuangbanna, Yunnan Province, China. Vector Borne Zoonotic Dis 2023. [PMID: 37184906 DOI: 10.1089/vbz.2022.0091] [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: 05/16/2023] Open
Abstract
Background: Dengue virus (DENV) can be divided into four serotypes-DENV-1, DENV-2, DENV-3, and DENV-4. In humans, infection leads to dengue fever (DF), dengue hemorrhagic fever, and dengue shock syndrome, both widely prevalent in tropical and subtropical regions. In 2019, a severe outbreak of DF occurred in Xishuangbanna, Yunnan province. Objective: To investigate the etiology and genotype of the causative agents of this severe dengue outbreak in Xishuangbanna. Methods: Between October and November 2019, the sera of patients clinically diagnosed with DF were collected in the first People's Hospital of Xishuangbanna. RNA was extracted from the sera and amplified by RT-PCR with flavivirus primers. Flavivirus-positive sera were then used to inoculate Aedes albopictus cells (C6/36); viral RNA was extracted from these cells, amplified, and sequenced with DENV E gene-specific primers. Sequence splicing and nucleotide homology genetic evolution analysis were carried out by biological software (DNAStar). Unique mutations in the E genes of isolated DENV were analyzed by SWISS-MODEL and PyMOL. Results: Of the 60 samples collected from DF patients, 39 tested positively with flavivirus primers. The DENV was isolated from 25 of the 39 positive seras, of which 20 showed cytopathic effects (CPE) and 5 were no CPE. In these 25 isolated nucleic acids, 21 strains of DENV-1, 3 strains of DENV-2, and 1 strain of DENV-3 were identified according to the sequence of E protein. In the four unique mutations (D52, Y149, L312, T386), D52 and Y149 in the E protein of DENV-1 were predicted to be exposed on the surface of the prefusion conformation. Conclusion: The 2019 outbreak of DF in Xishuangbanna area of Yunnan Province consists of at least three serotypes of DENV-1, DENV-2, and DENV-3, and the sources of these virus strains are of mixed and complicated origin.
Collapse
Affiliation(s)
- Jin-Xin Meng
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Qiu-Ming Hu
- People's Hospital of Jinghong City, Jinghong, China
| | - Li-Ming Zhang
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Nan Li
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Yu-Wen He
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Zhen-Xing Yang
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming, China
| | - Yi Sun
- The First People's Hospital of Yunnan Province, Kunming, China
| | - Jing-Lin Wang
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Animal Science and Veterinary Institute, Kunming, China
| |
Collapse
|
5
|
Lee MF, Anasir MI, Poh CL. Development of novel antiviral peptides against dengue serotypes 1-4. Virology 2023; 580:10-27. [PMID: 36739680 DOI: 10.1016/j.virol.2023.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/03/2023]
Abstract
Dengue infections pose a critical threat to public health worldwide. Since there are no clinically approved antiviral drugs to treat dengue infections caused by the four dengue virus (DENV) serotypes, there is an urgent need to develop effective antivirals. Peptides are promising antiviral candidates due to their specificity and non-toxic properties. The DENV envelope (E) protein was selected for the design of antiviral peptides due to its importance in receptor binding and viral fusion to the host cell membrane. Twelve novel peptides were designed to mimic regions containing critical amino acid residues of the DENV E protein required for interaction with the host. A total of four peptides were identified to exhibit potent inhibitory effects against at least three or all four DENV serotypes. Peptide 3 demonstrated all three modes of action: cell protection and inhibition of post-infection against all four DENV serotypes, whereas direct virus-inactivating effects were only observed against DENV-2, 3, and 4. Peptide 4 showed good direct virus-inactivating effects against DENV-2 (74.26%) as well as good inhibitions of DENV-1 (80.37%) and DENV-4 (72.22%) during the post-infection stage. Peptide 5 exhibited direct virus-inactivating effects against all four DENV serotypes, albeit at lower inhibition levels against DENV-1 and DENV-3. It also exhibited highly significant inhibition of DENV-4 (89.31%) during post-infection. Truncated peptide 5F which was derived from peptide 5 showed more significant inhibition of DENV-4 (91.58%) during post-infection and good direct virus-inactivating effects against DENV-2 (77.55%) at a lower concentration of 100 μM. Peptide 3 could be considered as the best antiviral candidate for pre- and post-infection treatments of DENV infections in regions with four circulating dengue serotypes. However, if the most predominant dengue serotype for a particular region could be identified, peptides with significantly high antiviral activities against that particular dengue serotype could serve as more suitable antiviral candidates. Thus, peptide 5F serves as a more suitable antiviral candidate for post-infection treatment against DENV-4.
Collapse
Affiliation(s)
- Michelle Felicia Lee
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, 5, Jalan Universiti, Bandar Sunway, Selangor, 47500, Malaysia
| | - Mohd Ishtiaq Anasir
- Virology Unit, Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Setia Alam, Shah Alam, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, 5, Jalan Universiti, Bandar Sunway, Selangor, 47500, Malaysia.
| |
Collapse
|
6
|
Zerfu B, Kassa T, Legesse M. Epidemiology, biology, pathogenesis, clinical manifestations, and diagnosis of dengue virus infection, and its trend in Ethiopia: a comprehensive literature review. Trop Med Health 2023; 51:11. [PMID: 36829222 PMCID: PMC9950709 DOI: 10.1186/s41182-023-00504-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/15/2023] [Indexed: 02/26/2023] Open
Abstract
Dengue fever is a dengue virus infection, emerging rapidly and posing public health threat worldwide, primarily in tropical and subtropical countries. Nearly half of the world's population is now at risk of contracting the dengue virus, including new countries with no previous history-like Ethiopia. However, little is known about the epidemiology and impact of the disease in different countries. This is especially true in countries, where cases have recently begun to be reported. This review aims to summarize epidemiology, biology, pathogenesis, clinical manifestations, and diagnosis of dengue virus infection and its trend in Ethiopia. It may help countries, where dengue fever is not yet on the public health list-like Ethiopia to alert healthcare workers to consider the disease for diagnosis and treatment. The review retrieved and incorporated 139 published and organizational reports showing approximately 390 million new infections. About 100 million of these infections develop the clinical features of dengue, and thousands of people die annually from severe dengue fever in 129 countries. It is caused by being bitten by a dengue virus-infected female mosquito, primarily Aedes aegypti and, lesser, Ae. albopictus. Dengue virus is a member of the Flavivirus genus of the Flaviviridae family and has four independent but antigen-related single-stranded positive-sense RNA virus serotypes. The infection is usually asymptomatic but causes illnesses ranging from mild febrile illness to fatal dengue hemorrhagic fever or shock syndrome. Diagnosis can be by detecting the virus genome using nucleic acids amplification tests or testing NS1 antigen and/or anti-dengue antibodies from serum, plasma, circulating blood cells, or other tissues. Dengue cases and outbreaks have increased in recent decades, with a significant public health impact. Ethiopia has had nearly annual outbreaks since 2013, devastating an already fragmented health system and economy. Standardization of medication, population-level screening for early diagnosis and prompt treatment, and minimization of mosquito bites reduce overall infection and mortality rates.
Collapse
Affiliation(s)
- Biruk Zerfu
- Department of Medical Laboratory Science, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia. .,Aklilu Lema Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Tesfu Kassa
- grid.7123.70000 0001 1250 5688Aklilu Lema Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Mengistu Legesse
- grid.7123.70000 0001 1250 5688Aklilu Lema Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
7
|
Sun Y, Ding H, Zhao F, Yan Q, Li Y, Niu X, Zeng W, Wu K, Ling B, Fan S, Zhao M, Yi L, Chen J. Genomic Characteristics and E Protein Bioinformatics Analysis of JEV Isolates from South China from 2011 to 2018. Vaccines (Basel) 2022; 10:vaccines10081303. [PMID: 36016192 PMCID: PMC9412759 DOI: 10.3390/vaccines10081303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Japanese encephalitis is a mosquito-borne zoonotic epidemic caused by the Japanese encephalitis virus (JEV). JEV is not only the leading cause of Asian viral encephalitis, but also one of the leading causes of viral encephalitis worldwide. To understand the genetic evolution and E protein characteristics of JEV, 263 suspected porcine JE samples collected from South China from 2011 to 2018 were inspected. It was found that 78 aborted porcine fetuses were JEV-nucleic-acid-positive, with a positive rate of 29.7%. Furthermore, four JEV variants were isolated from JEV-nucleic-acid-positive materials, namely, CH/GD2011/2011, CH/GD2014/2014, CH/GD2015/2015, and CH/GD2018/2018. The cell culture and virus titer determination of four JEV isolates showed that four JEV isolates could proliferate stably in Vero cells, and the virus titer was as high as 108.5 TCID 50/mL. The whole-genome sequences of four JEV isolates were sequenced. Based on the phylogenetic analysis of the JEV E gene and whole genome, it was found that CH/GD2011/2011 and CH/GD2015/2015 belonged to the GIII type, while CH/GD2014/2014 and CH/GD2018/2018 belonged to the GI type, which was significantly different from that of the JEV classical strain CH/BJ-1/1995. Bioinformatics tools were used to analyze the E protein phosphorylation site, glycosylation site, B cell antigen epitope, and modeled 3D structures of E protein in four JEV isolates. The analysis of the prevalence of JEV and the biological function of E protein can provide a theoretical basis for the prevention and control of JEV and the design of antiviral drugs.
Collapse
Affiliation(s)
- Yawei Sun
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Hongxing Ding
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Feifan Zhao
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Quanhui Yan
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yuwan Li
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Xinni Niu
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Weijun Zeng
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Keke Wu
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Bing Ling
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shuangqi Fan
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Mingqiu Zhao
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Lin Yi
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Jinding Chen
- Department of Microbiology and Immunology, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: ; Fax: +86-20-8528-0245
| |
Collapse
|
8
|
Pfaff-Kilgore JM, Davidson E, Kadash-Edmondson K, Hernandez M, Rosenberg E, Chambers R, Castelli M, Clementi N, Mancini N, Bailey JR, Crowe JE, Law M, Doranz BJ. Sites of vulnerability in HCV E1E2 identified by comprehensive functional screening. Cell Rep 2022; 39:110859. [PMID: 35613596 PMCID: PMC9281441 DOI: 10.1016/j.celrep.2022.110859] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 12/08/2021] [Accepted: 05/01/2022] [Indexed: 12/15/2022] Open
Abstract
The E1 and E2 envelope proteins of hepatitis C virus (HCV) form a heterodimer that drives virus-host membrane fusion. Here, we analyze the role of each amino acid in E1E2 function, expressing 545 individual alanine mutants of E1E2 in human cells, incorporating them into infectious viral pseudoparticles, and testing them against 37 different monoclonal antibodies (MAbs) to ascertain full-length translation, folding, heterodimer assembly, CD81 binding, viral pseudoparticle incorporation, and infectivity. We propose a model describing the role of each critical residue in E1E2 functionality and use it to examine how MAbs neutralize infection by exploiting functionally critical sites of vulnerability on E1E2. Our results suggest that E1E2 is a surprisingly fragile protein complex where even a single alanine mutation at 92% of positions disrupts its function. The amino-acid-level targets identified are highly conserved and functionally critical and can be exploited for improved therapies and vaccines.
Collapse
Affiliation(s)
| | - Edgar Davidson
- Integral Molecular, Inc., 3711 Market St, Philadelphia, PA 19104, USA
| | | | - Mayda Hernandez
- Integral Molecular, Inc., 3711 Market St, Philadelphia, PA 19104, USA
| | - Erin Rosenberg
- Integral Molecular, Inc., 3711 Market St, Philadelphia, PA 19104, USA
| | - Ross Chambers
- Integral Molecular, Inc., 3711 Market St, Philadelphia, PA 19104, USA
| | - Matteo Castelli
- Laboratory of Medical Microbiology and Virology, University Vita-Salute San Raffaele, Milan, Italy
| | - Nicola Clementi
- Laboratory of Medical Microbiology and Virology, University Vita-Salute San Raffaele, Milan, Italy; IRCSS San Raffaele Hospital, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, University Vita-Salute San Raffaele, Milan, Italy; IRCSS San Raffaele Hospital, Milan, Italy
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mansun Law
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Benjamin J Doranz
- Integral Molecular, Inc., 3711 Market St, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Differential heterologous neutralisation profile against strains within DENV-3 genotype II. Epidemiol Infect 2021; 150:e33. [PMID: 35225194 PMCID: PMC8888274 DOI: 10.1017/s0950268821002648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The dengue virus type 3 (DENV-3) homotypic outbreak cycles reported in Klang Valley, Malaysia in 1992–1995 and 2002 demonstrated different epidemic magnitude and duration. These outbreak cycles were caused by two closely related strains of viruses within the DENV-3 genotype II (DENV-3/II). The role of viral genotypic diversity and factors that could have influenced this phenomenon were investigated. The serum neutralisation sensitivity of DEN3/II strains responsible for the DENV-3 outbreak cycles in 1992–1995 and 2002 were examined. Representative virus isolates from the respective outbreaks were subjected to virus neutralisation assay using identified sera of patients with homotypic (DENV-3) or heterotypic dengue infections (DENV-1 and DENV-2). Results from the study suggested that isolates representing DENV-3/II group E (DENV-3/II-E) from the 1992–1995 outbreak and DENV-3/II group F (DENV-3/II-F) from the 2002 outbreak were neutralised at similar capacity (intergenotypic differences <2-fold) by sera of patients infected with DENV-3, DENV-1 and DENV-2/Asian genotypes. Sera of the DENV-2/Cosmopolitan infection efficiently neutralised DENV-3/II-F (FRNT50 = 508.0) at a similar neutralisation capacity against its own homotypic serotype, DENV-2 (FRNT50 = 452.5), but not against DENV-3/II-E (FRNT50 = 100.8). The different neutralisation sensitivities of DENV-3/II strains towards the cross-reacting DENV-2 heterotypic immunity could play a role in shaping the DENV-3 recurring outbreaks pattern in Malaysia. Two genetic variations, E-132 (H/Y) and E-479 (A/V) were identified on the envelope protein of DENV-3/II-E and DENV-3/II-F, respectively. The E-132 variation was predicted to affect the protein stability. A more extensive study, however, on the implication of the naturally occurring genetic variations within closely related DENV genotypes on the neutralisation profile and protective immunity would be needed for a better understanding of the DENV spread pattern in a hyperendemic setting.
Collapse
|
10
|
Düzgüneş N, Fernandez-Fuentes N, Konopka K. Inhibition of Viral Membrane Fusion by Peptides and Approaches to Peptide Design. Pathogens 2021; 10:1599. [PMID: 34959554 PMCID: PMC8709411 DOI: 10.3390/pathogens10121599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Fusion of lipid-enveloped viruses with the cellular plasma membrane or the endosome membrane is mediated by viral envelope proteins that undergo large conformational changes following binding to receptors. The HIV-1 fusion protein gp41 undergoes a transition into a "six-helix bundle" after binding of the surface protein gp120 to the CD4 receptor and a co-receptor. Synthetic peptides that mimic part of this structure interfere with the formation of the helix structure and inhibit membrane fusion. This approach also works with the S spike protein of SARS-CoV-2. Here we review the peptide inhibitors of membrane fusion involved in infection by influenza virus, HIV-1, MERS and SARS coronaviruses, hepatitis viruses, paramyxoviruses, flaviviruses, herpesviruses and filoviruses. We also describe recent computational methods used for the identification of peptide sequences that can interact strongly with protein interfaces, with special emphasis on SARS-CoV-2, using the PePI-Covid19 database.
Collapse
Affiliation(s)
- Nejat Düzgüneş
- Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA;
| | - Narcis Fernandez-Fuentes
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth SY23 3EE, UK;
| | - Krystyna Konopka
- Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103, USA;
| |
Collapse
|
11
|
Gavor E, Choong YK, Tulsian NK, Nayak D, Idris F, Sivaraman H, Ting DHR, Sylvie A, Mok YK, Kini RM, Sivaraman J. Structure of Aedes aegypti procarboxypeptidase B1 and its binding with Dengue virus for controlling infection. Life Sci Alliance 2021; 5:5/1/e202101211. [PMID: 34750241 PMCID: PMC8605224 DOI: 10.26508/lsa.202101211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/05/2022] Open
Abstract
Using high-resolution structure, we have characterized the substrate specificity of Aedes aegypti procarboxypeptidase B1 and provided mechanistic insights into the binding and inhibition of DENV Metallocarboxypeptidases play critical roles in the development of mosquitoes and influence pathogen/parasite infection of the mosquito midgut. Here, we report the crystal structure of Aedes aegypti procarboxypeptidase B1 (PCPBAe1), characterized its substrate specificity and mechanism of binding to and inhibiting Dengue virus (DENV). We show that the activated PCPBAe1 (CPBAe1) hydrolyzes both Arg- and Lys-substrates, which is modulated by residues Asp251 and Ser239. Notably, these residues are conserved in CPBs across mosquito species, possibly required for efficient digestion of basic dietary residues that are necessary for mosquito reproduction and development. Importantly, we characterized the interaction between PCPBAe1 and DENV envelope (E) protein, virus-like particles, and infectious virions. We identified residues Asp18A, Glu19A, Glu85, Arg87, and Arg89 of PCPBAe1 are essential for interaction with DENV. PCPBAe1 maps to the dimeric interface of the E protein domains I/II (Lys64–Glu84, Val238–Val252, and Leu278–Leu287). Overall, our studies provide general insights into how the substrate-binding property of mosquito carboxypeptidases could be targeted to potentially control mosquito populations or proposes a mechanism by which PCPBAe1 binds to and inhibits DENV.
Collapse
Affiliation(s)
- Edem Gavor
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yeu Khai Choong
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Nikhil Kumar Tulsian
- Department of Biological Sciences, National University of Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Digant Nayak
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Fakhriedzwan Idris
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Hariharan Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Donald Heng Rong Ting
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Alonso Sylvie
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Yu Keung Mok
- Department of Biological Sciences, National University of Singapore, Singapore
| | - R Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
12
|
Low ZX, OuYong BM, Hassandarvish P, Poh CL, Ramanathan B. Antiviral activity of silymarin and baicalein against dengue virus. Sci Rep 2021; 11:21221. [PMID: 34707245 PMCID: PMC8551334 DOI: 10.1038/s41598-021-98949-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Dengue is an arthropod-borne viral disease that has become endemic and a global threat in many countries with no effective antiviral drug available currently. This study showed that flavonoids: silymarin and baicalein could inhibit the dengue virus in vitro and were well tolerated in Vero cells with a half-maximum cytotoxic concentration (CC50) of 749.70 µg/mL and 271.03 µg/mL, respectively. Silymarin and baicalein exerted virucidal effects against DENV-3, with a selective index (SI) of 10.87 and 21.34, respectively. Baicalein showed a better inhibition of intracellular DENV-3 progeny with a SI of 7.82 compared to silymarin. Baicalein effectively blocked DENV-3 attachment (95.59%) to the Vero cells, while silymarin prevented the viral entry (72.46%) into the cells, thus reducing viral infectivity. Both flavonoids showed promising antiviral activity against all four dengue serotypes. The in silico molecular docking showed that silymarin could bind to the viral envelope (E) protein with a binding affinity of - 8.5 kcal/mol and form hydrogen bonds with the amino acids GLN120, TRP229, ASN89, and THR223 of the E protein. Overall, this study showed that silymarin and baicalein exhibited potential anti-DENV activity and could serve as promising antiviral agents for further development against dengue infection.
Collapse
Affiliation(s)
- Zhao Xuan Low
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Kuala Lumpur, Malaysia
| | - Brian Ming OuYong
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Kuala Lumpur, Malaysia
| | - Pouya Hassandarvish
- Tropical Infectious Diseases Research and Education Centre, University Malaya, 50603, Kuala Lumpur, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Kuala Lumpur, Malaysia
| | - Babu Ramanathan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Kuala Lumpur, Malaysia.
| |
Collapse
|
13
|
Kudlacek ST, Metz S, Thiono D, Payne AM, Phan TTN, Tian S, Forsberg LJ, Maguire J, Seim I, Zhang S, Tripathy A, Harrison J, Nicely NI, Soman S, McCracken MK, Gromowski GD, Jarman RG, Premkumar L, de Silva AM, Kuhlman B. Designed, highly expressing, thermostable dengue virus 2 envelope protein dimers elicit quaternary epitope antibodies. SCIENCE ADVANCES 2021; 7:eabg4084. [PMID: 34652943 PMCID: PMC8519570 DOI: 10.1126/sciadv.abg4084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/25/2021] [Indexed: 05/30/2023]
Abstract
Dengue virus (DENV) is a worldwide health burden, and a safe vaccine is needed. Neutralizing antibodies bind to quaternary epitopes on DENV envelope (E) protein homodimers. However, recombinantly expressed soluble E proteins are monomers under vaccination conditions and do not present these quaternary epitopes, partly explaining their limited success as vaccine antigens. Using molecular modeling, we found DENV2 E protein mutations that induce dimerization at low concentrations (<100 pM) and enhance production yield by more than 50-fold. Cross-dimer epitope antibodies bind to the stabilized dimers, and a crystal structure resembles the wild-type (WT) E protein bound to a dimer epitope antibody. Mice immunized with the stabilized dimers developed antibodies that bind to E dimers and not monomers and elicited higher levels of DENV2-neutralizing antibodies compared to mice immunized with WT E antigen. Our findings demonstrate the feasibility of using structure-based design to produce subunit vaccines for dengue and other flaviviruses.
Collapse
Affiliation(s)
- Stephan T. Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Stefan Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Devina Thiono
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Alexander M. Payne
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Thanh T. N. Phan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Lawrence J. Forsberg
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Jack Maguire
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ian Seim
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shu Zhang
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ashutosh Tripathy
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Joseph Harrison
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Nathan I. Nicely
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Sandrine Soman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Michael K. McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory D. Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Richard G. Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
| |
Collapse
|
14
|
Wang WH, Urbina AN, Lin CY, Yang ZS, Assavalapsakul W, Thitithanyanont A, Lu PL, Chen YH, Wang SF. Targets and strategies for vaccine development against dengue viruses. Biomed Pharmacother 2021; 144:112304. [PMID: 34634560 DOI: 10.1016/j.biopha.2021.112304] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 10/20/2022] Open
Abstract
Dengue virus (DENV) is a global health threat causing about half of the worldwide population to be at risk of infection, especially the people living in tropical and subtropical area. Although the dengue disease caused by dengue virus (DENV) is asymptomatic and self-limiting in most people with first infection, increased severe dengue symptoms may be observed in people with heterotypic secondary DENV infection. Since there is a lack of specific antiviral medication, the development of dengue vaccines is critical in the prevention and control this disease. Several targets and strategies in the development of dengue vaccine have been demonstrated. Currently, Dengvaxia, a live-attenuated chimeric yellow-fever/tetravalent dengue vaccine (CYD-TDV) developed by Sanofi Pasteur, has been licensed and approved for clinical use in some countries. However, this vaccine has demonstrated low efficacy in children and dengue-naïve individuals and also increases the risk of severe dengue in young vaccinated recipients. Accordingly, many novel strategies for the dengue vaccine are under investigation and development. Here, we conducted a systemic literature review according to PRISMA guidelines to give a concise overview of various aspects of the vaccine development process against DENVs, mainly targeting five potential strategies including live attenuated vaccine, inactivated virus vaccine, recombinant subunit vaccine, viral-vector vaccine, and DNA vaccine. This study offers the comprehensive view of updated information and current progression of immunogen selection as well as strategies of vaccine development against DENVs.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Aspiro Nayim Urbina
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chih-Yen Lin
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Zih-Syuan Yang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Po-Liang Lu
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Yen-Hsu Chen
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical, University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
15
|
Imagawa T, Ito M, Matsuda M, Nakashima K, Tokunaga Y, Ohta I, Li TC, Suzuki R, Suzuki T. Virus-like particles with FLAG-tagged envelope protein as a tetravalent dengue vaccine candidate. Sci Rep 2021; 11:17542. [PMID: 34475493 PMCID: PMC8413300 DOI: 10.1038/s41598-021-97038-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
The global incidence of dengue, which is caused by dengue virus (DENV) infection, has grown dramatically in recent decades and secondary infection with heterologous serotype of the virus may cause severe symptoms. Efficacious dengue vaccines should be able to provide long-lasting immunity against all four DENV serotypes simultaneously. In this study, we constructed a novel vaccine platform based on tetravalent dengue virus-like particles (DENV-LPs) in which envelope (E) protein carried a FLAG tag sequence at the position located not only in the exterior loop on the protruding domain but outside of dimerization interface of the protein. We demonstrated an effective strategy to produce the DENV-LPs by transient transfection with expression plasmids for pre-membrane and E proteins of DENV-1 to DENV-4 in mammalian cells and to concentrate and purify them with one-step affinity chromatography. Characteristic features of VLPs such as particle size, shape and density were comparable to flavivirus-like particles reported. The neutralizing activity against all four DENV serotypes was successfully induced by immunization with the purified tetravalent VLPs in mice. Simple, one-step purification systems for VLP vaccine platforms using epitope-tagging strategy should be advantageous for vaccine development not only for dengue but for emerging pandemics in the future.
Collapse
Affiliation(s)
- Toshifumi Imagawa
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Masahiko Ito
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Mami Matsuda
- Department of Virology II, National Institute for Infectious Disease, Musashi-murayama, Tokyo, 208-0011, Japan
| | - Kenji Nakashima
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Yuhei Tokunaga
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Isao Ohta
- Advanced Research Facilities and Services, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan
| | - Tian-Cheng Li
- Department of Virology II, National Institute for Infectious Disease, Musashi-murayama, Tokyo, 208-0011, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute for Infectious Disease, Musashi-murayama, Tokyo, 208-0011, Japan
| | - Tetsuro Suzuki
- Department of Virology and Parasitology, Hamamatsu University School of Medicine, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
16
|
S AH, Pujar GV, Sethu AK, Bhagyalalitha M, Singh M. Dengue structural proteins as antiviral drug targets: Current status in the drug discovery & development. Eur J Med Chem 2021; 221:113527. [PMID: 34020338 DOI: 10.1016/j.ejmech.2021.113527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/30/2021] [Accepted: 04/20/2021] [Indexed: 01/04/2023]
Abstract
Dengue virus belongs to the class of RNA viruses and subclass of enveloped single-stranded positive-sense RNA virus. It causes dengue fever (DF), dengue hemorrhagic fever (DHF), or dengue shock syndrome (DSS), where DHF and DSS are life-threatening. Even though dengue is an age-old disease, it is still a mystery and continues to be a global threat. Numerous attempts have been carried out in the past few decades to eradicate the virus through vaccine and antiviral drugs, but still battle continues. In this review, the possible drug targets for discovery and development of potential antiviral drugs against structural proteins of dengue virus, the current development status of the antiviral drugs against dengue around the world, and challenges that need to be addressed to overcome the shortcomings in the process of drug discovery have been discussed.
Collapse
Affiliation(s)
- Akshatha H S
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015, India
| | - Gurubasavaraj V Pujar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015, India.
| | - Arun Kumar Sethu
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015, India
| | - Meduri Bhagyalalitha
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015, India
| | - Manisha Singh
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Sri Shivarathreeshwara Nagara, Mysuru, 570015, India
| |
Collapse
|
17
|
Whitbeck JC, Thomas A, Kadash-Edmondson K, Grinyo-Escuer A, Stafford LJ, Cheng C, Liao GC, Holtsberg FW, Aman MJ, Simmons G, Davidson E, Doranz BJ. Antigenicity, stability, and reproducibility of Zika reporter virus particles for long-term applications. PLoS Negl Trop Dis 2020; 14:e0008730. [PMID: 33206639 PMCID: PMC7673510 DOI: 10.1371/journal.pntd.0008730] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/17/2020] [Indexed: 01/16/2023] Open
Abstract
The development of vaccines against flaviviruses, including Zika virus (ZIKV) and dengue virus (DENV), continues to be a major challenge, hindered by the lack of efficient and reliable methods for screening neutralizing activity of sera or antibodies. To address this need, we previously developed a plasmid-based, replication-incompetent DENV reporter virus particle (RVP) production system as an efficient and safe alternative to the Plaque Reduction Neutralization Test (PRNT). As part of the response to the 2015-2016 ZIKV outbreak, we developed pseudo-infectious ZIKV RVPs by modifying our DENV RVP system. The use of ZIKV RVPs as critical reagents in human clinical trials requires their further validation using stability and reproducibility metrics for large-scale applications. In the current study, we validated ZIKV RVPs using infectivity, neutralization, and enhancement assays with monoclonal antibodies (MAbs) and human ZIKV-positive patient serum. ZIKV RVPs are antigenically equivalent to live virus based on binding ELISA and neutralization results and are nonreplicating based on the results of live virus replication assays. We demonstrate reproducible neutralization titer data (NT50 values) across different RVP production lots, volumes, time frames, and laboratories. We also show RVP stability across experimentally relevant time intervals and temperatures. Our results demonstrate that ZIKV RVPs provide a safe, high-throughput, and reproducible reagent for large-scale, long-term studies of neutralizing antibodies and sera, which can facilitate large-scale screening and epidemiological studies to help expedite ZIKV vaccine development.
Collapse
Affiliation(s)
| | - Anu Thomas
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | | | | | - Lewis J. Stafford
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | - Celine Cheng
- Vitalant Research Institute, San Francisco, CA, United States of America
| | - Grant C. Liao
- Integrated Biotherapeutics, 4 Research Court, Rockville, MD, United States of America
| | | | - M. Javad Aman
- Integrated Biotherapeutics, 4 Research Court, Rockville, MD, United States of America
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, CA, United States of America
| | - Edgar Davidson
- Integral Molecular, Inc., Philadelphia, PA, United States of America
| | | |
Collapse
|
18
|
Sharma N, Prosser O, Kumar P, Tuplin A, Giri R. Small molecule inhibitors possibly targeting the rearrangement of Zika virus envelope protein. Antiviral Res 2020; 182:104876. [PMID: 32783901 DOI: 10.1016/j.antiviral.2020.104876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/12/2023]
Abstract
The recurrent public health threat imposed by Zika Virus (ZIKV) in various geographical areas necessitates the immediate development of antiviral compounds or vaccines. Flaviviral Envelope (E) proteins are essential for host-cell recognition and virion entry. Consequently, they represent an important target for antiviral therapy, with the aim of preventing viral spread during early stages of infection. Due to conformational rearrangement during entry, flavivirus E proteins present several alternative conformations as potential antiviral targets - for blocking entry or virus-host membrane fusion. We previously identified a conserved hydrophobic region, between DI/DIII of ZIKV E protein, with potential to act as an antiviral target. Here, we screened commercially available antiviral compound libraries against ZIKV E protein, using a structure-based drug discovery approach. The antiviral efficacy of the top ten screened compounds were experimentally validated for inhibition of ZIKV replication in Vero Cells. Compound F1065-0358 was observed to inhibit ZIKV replication with an IC50 of 14.0 μM. Ligand-protein complex molecular dynamic simulations confirmed the stability of ligand binding up to 100 ns. Together, results from this study indicate that F1065-0358 functions as a ZIKV virus inhibitor by interfering E protein conformational rearrangement. Furthermore, given that F1065-0358 interacts with highly conserved residues of E protein, this raises the potential for its efficacy against other pathogenic flaviviruses.
Collapse
Affiliation(s)
- Nitin Sharma
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Oliver Prosser
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Andrew Tuplin
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India.
| |
Collapse
|
19
|
Versiani AF, Martins EMN, Andrade LM, Cox L, Pereira GC, Barbosa-Stancioli EF, Nogueira ML, Ladeira LO, da Fonseca FG. Nanosensors based on LSPR are able to serologically differentiate dengue from Zika infections. Sci Rep 2020; 10:11302. [PMID: 32647259 PMCID: PMC7347616 DOI: 10.1038/s41598-020-68357-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/22/2020] [Indexed: 01/16/2023] Open
Abstract
The Flaviviridae virus family was named after the Yellow-fever virus, and the latin term flavi means “of golden color”. Dengue, caused by Dengue virus (DENV), is one of the most important infectious diseases worldwide. A sensitive and differential diagnosis is crucial for patient management, especially due to the occurrence of serological cross-reactivity to other co-circulating flaviviruses. This became particularly important with the emergence of Zika virus (ZIKV) in areas were DENV seroprevalence was already high. We developed a sensitive and specific diagnostic test based on gold nanorods (GNR) functionalized with DENV proteins as nanosensors. These were able to detect as little as one picogram of anti-DENV monoclonal antibodies and highly diluted DENV-positive human sera. The nanosensors could differentiate DENV-positive sera from other flavivirus-infected patients, including ZIKV, and were even able to distinguish which DENV serotype infected individual patients. Readouts are obtained in ELISA-plate spectrophotometers without the need of specific devices.
Collapse
Affiliation(s)
- Alice F Versiani
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Laboratório de Pesquisa em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José Do Rio Preto, São José do Rio Preto, SP, Brazil
| | - Estefânia M N Martins
- Laboratório de Química de Nanoestruturas de Carbono, Centro de Desenvolvimento da Tecnologia Nuclear-CDTN/CNEN, Belo Horizonte, MG, Brazil.,NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lidia M Andrade
- Laboratório de Nanomateriais, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Laura Cox
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Edel F Barbosa-Stancioli
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauricio L Nogueira
- Laboratório de Pesquisa em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José Do Rio Preto, São José do Rio Preto, SP, Brazil
| | - Luiz O Ladeira
- Laboratório de Nanomateriais, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,NanoBioMedical Research Group, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávio G da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil. .,Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
20
|
Sauer MF, Sevy AM, Crowe JE, Meiler J. Multi-state design of flexible proteins predicts sequences optimal for conformational change. PLoS Comput Biol 2020; 16:e1007339. [PMID: 32032348 PMCID: PMC7032724 DOI: 10.1371/journal.pcbi.1007339] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/20/2020] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Computational protein design of an ensemble of conformations for one protein–i.e., multi-state design–determines the side chain identity by optimizing the energetic contributions of that side chain in each of the backbone conformations. Sampling the resulting large sequence-structure search space limits the number of conformations and the size of proteins in multi-state design algorithms. Here, we demonstrated that the REstrained CONvergence (RECON) algorithm can simultaneously evaluate the sequence of large proteins that undergo substantial conformational changes. Simultaneous optimization of side chain conformations across all conformations increased sequence conservation when compared to single-state designs in all cases. More importantly, the sequence space sampled by RECON MSD resembled the evolutionary sequence space of flexible proteins, particularly when confined to predicting the mutational preferences of limited common ancestral descent, such as in the case of influenza type A hemagglutinin. Additionally, we found that sequence positions which require substantial changes in their local environment across an ensemble of conformations are more likely to be conserved. These increased conservation rates are better captured by RECON MSD over multiple conformations and thus multiple local residue environments during design. To quantify this rewiring of contacts at a certain position in sequence and structure, we introduced a new metric designated ‘contact proximity deviation’ that enumerates contact map changes. This measure allows mapping of global conformational changes into local side chain proximity adjustments, a property not captured by traditional global similarity metrics such as RMSD or local similarity metrics such as changes in φ and ψ angles. Multi-state design can be used to engineer proteins that need to exist in multiple conformations or that bind to multiple partner molecules. In essence, multi-state design selects a compromise of protein sequences that allow for an ensemble of protein conformations, or states, associated with a particular biological function. In this paper, we used the REstrained CONvergence (RECON) algorithm with Rosetta to show that multi-state design of flexible proteins predicts sequences optimal for conformational change, mimicking mutation preferences sampled in evolution. Modeling optimal local side chain physicochemical environments within an ensemble selected significantly more native-like sequences than selections performed when all conformations states are designed independently. This outcome was particularly true for amino acids whose local side chain environment change between conformations. To quantify such contact map changes, we introduced a novel metric to show that sequence conservation is dependent on protein flexibility, i.e., changes in local side chain environments between stated limit the space of tolerated mutations. Additionally, such positions in sequence and structure are more likely to be energetically frustrated, at least in some states. Importantly, we showed that multi-state design over an ensemble of conformations (space) can explore evolutionary tolerated sequence space (time), thus enabling RECON to not only design proteins that require multiple states for function but also predict mutations that might be tolerated in native proteins but have not yet been explored by evolution. The latter aspect can be important to anticipate escape mutations, for example in pathogens or oncoproteins.
Collapse
Affiliation(s)
- Marion F Sauer
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America.,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexander M Sevy
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America.,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America.,Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
21
|
Dengue virus envelope protein domain III induces pro-inflammatory signature and triggers activation of inflammasome. Cytokine 2019; 123:154780. [DOI: 10.1016/j.cyto.2019.154780] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 06/30/2019] [Accepted: 07/09/2019] [Indexed: 01/03/2023]
|
22
|
Li L, Meng W, Horton M, DiStefano DR, Thoryk EA, Pfaff JM, Wang Q, Salazar GT, Barnes T, Doranz BJ, Bett AJ, Casimiro DR, Vora KA, An Z, Zhang N. Potent neutralizing antibodies elicited by dengue vaccine in rhesus macaque target diverse epitopes. PLoS Pathog 2019; 15:e1007716. [PMID: 31170257 PMCID: PMC6553876 DOI: 10.1371/journal.ppat.1007716] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/19/2019] [Indexed: 01/11/2023] Open
Abstract
There is still no safe and effective vaccine against dengue virus infection. Epidemics of dengue virus infection are increasingly a threat to human health around the world. Antibodies generated in response to dengue infection have been shown to impact disease development and effectiveness of dengue vaccine. In this study, we investigated monoclonal antibody responses to an experimental dengue vaccine in rhesus macaques. Variable regions of both heavy chain (VH) and light chain (VL) were cloned from single antibody-secreting B cells. A total of 780 monoclonal antibodies (mAbs) composed of paired VH and VL were characterized. Results show that the vaccination induces mAbs with diverse germline sequences and a wide range of binding affinities. Six potent neutralizing mAbs were identified among 130 dengue envelope protein binders. Critical amino acids for each neutralizing antibody binding to the dengue envelope protein were identified by alanine scanning of mutant libraries. Diverse epitopes were identified, including epitopes on the lateral ridge of DIII, the I-III hinge, the bc loop adjacent to the fusion loop of DII, and the β-strands and loops of DI. Significantly, one of the neutralizing mAbs has a previously unknown epitope in DII at the interface of the envelope and membrane protein and is capable of neutralizing all four dengue serotypes. Taken together, the results of this study not only provide preclinical validation for the tested experimental vaccine, but also shed light on a potential application of the rhesus macaque model for better dengue vaccine evaluation and design of vaccines and immunization strategies. Dengue virus (DENV) is a leading cause of human illness in the tropics and subtropics, with about 40% of the world’s population living in areas at risk for infection. There are four DENV serotypes. Patients who have previously been infected by one dengue serotype may develop more severe symptoms such as bleeding and endothelial leakage upon secondary infection with another dengue serotype. This study reports the extensive cloning and analysis of 780 monoclonal antibodies (mAbs) from single B cells of rhesus macaques after immunization with an experimental dengue vaccine. We identified a panel of potent neutralizing mAbs with diverse epitopes on the DENV envelope protein. Antibodies in this panel were found to bind to the lateral ridge of DIII, the I-III hinge, the bc loop adjacent to the fusion loop of DII, and the β-strands and the loops of DI. We also isolated one mAb (d448) that can neutralize all four dengue serotypes and binds to a novel epitope at the interface of the DENV envelope and membrane proteins. Further investigation of these neutralizing monoclonal antibodies is warranted for better vaccine efficacy evaluation and vaccine design.
Collapse
Affiliation(s)
- Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Weixu Meng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Melanie Horton
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
| | - Daniel R. DiStefano
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
| | - Elizabeth A. Thoryk
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
| | - Jennifer M. Pfaff
- Integral Molecular, Philadelphia, Pennsylvania, United States of America
| | - Qihui Wang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Georgina T. Salazar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Trevor Barnes
- Integral Molecular, Philadelphia, Pennsylvania, United States of America
| | - Benjamin J. Doranz
- Integral Molecular, Philadelphia, Pennsylvania, United States of America
| | - Andrew J. Bett
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
| | - Danilo R. Casimiro
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
| | - Kalpit A. Vora
- Department of Infectious Diseases and Vaccines Research, Merck Research Laboratories, Merck and Co. Inc., Kenilworth, New Jersey, United States of America
- * E-mail: (KV); (ZA); (NZ)
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail: (KV); (ZA); (NZ)
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail: (KV); (ZA); (NZ)
| |
Collapse
|
23
|
Ye C, Bian P, Zhang J, Xiao H, Zhang L, Ye W, Dong Y, Zhou Y, Jia Z, Lei Y. Structure-based discovery of antiviral inhibitors targeting the E dimer interface of Japanese encephalitis virus. Biochem Biophys Res Commun 2019; 515:366-371. [PMID: 31155294 DOI: 10.1016/j.bbrc.2019.05.148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 05/24/2019] [Indexed: 10/26/2022]
Abstract
Flaviviruses are emerging arthropod-borne viruses posing a great threat to human beings worldwide. The E dimer configuration of the flavivirus was prominent during viral assembly, maturation and entry. Neutralization antibodies targeting E dimer played the important role in controlling the flavivirus infection. Previously, the ideal drug target of small molecular inhibitors of JEV was viral proteases and polymerases. The crystal structure of JEV E protein showed a conserved pocket in it is important at membrane fusion step. Recently, a set of anti-virus drugs has been found by virtual screening. Here, we show that the fusion-loop pocket of JEV E protein was a conservative region and an ideal drug target. ChemDiv-3 from virtual screening as the lead compound was found to show a relatively modest inhibition effect for JEV in vitro and in vivo test and could interfere with the formation of JEV sE dimer. ChemDiv-3 interacts with the amino acid residues ASN 313, PRO 314, ALA 315, and VAL 323 in E protein via hydrogen bonds for occupation of the fusion-loop pocket. The key binding sites LYS 312, ALA 513 and THR 317 forming the fusion-loop pocket are the same and other auxiliary sites are similar among the flavivirus. Taken together, the fusion-loop pocket of the flavivirus could be one promising target for drug discovery.
Collapse
Affiliation(s)
- Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Peiyu Bian
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Han Xiao
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Li Zhang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Ye
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Yangchao Dong
- Department of Microbiology, Fourth Military Medical University, Xi'an, China
| | - Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| | - Yingfeng Lei
- Department of Microbiology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
24
|
Sabetian S, Nezafat N, Dorosti H, Zarei M, Ghasemi Y. Exploring dengue proteome to design an effective epitope-based vaccine against dengue virus. J Biomol Struct Dyn 2018; 37:2546-2563. [PMID: 30035699 DOI: 10.1080/07391102.2018.1491890] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dengue, a mosquito-borne disease, is caused by four known dengue serotypes. This infection causes a range of symptoms from a mild fever to a sever homorganic fever and death. It is a serious public health problem in subtropical and tropical countries. There is no specific vaccine currently available for clinical use and study on this issue is ongoing. In this study, bioinformatics approaches were used to predict antigenic, immunogenic, non-allergenic, and conserved B and T-cell epitopes as promising targets to design an effective peptide-based vaccine against dengue virus. Molecular docking analysis indicated the deep binding of the identified epitopes in the binding groove of the most popular human MHC I allele (human leukocyte antigens [HLA] A*0201). The final vaccine construct was created by conjugating the B and T-cell identified epitopes using proper linkers and adding an appropriate adjuvant at the N-terminal. The characteristics of the new subunit vaccine demonstrated that the epitope-based vaccine was antigenic, non-toxic, stable, and soluble. Other physicochemical properties of the new designed construct including isoelectric point value, aliphatic index, and grand average of hydropathicity were biologically considerable. Molecular docking of the engineered vaccine with Toll-like receptor 2 (TLR2) model revealed the hydrophobic interaction between the adjuvant and the ligand binding regions in the hydrophobic channel of TLR2. The study results indicated the high potential capability of the new multi-epitope vaccine to induce cellular and humoral immune responses against the dengue virus. Further experimental tests are required to investigate the immune protection capacity of the new vaccine construct in animal models. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Soudabeh Sabetian
- a Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Navid Nezafat
- a Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences , Shiraz , Iran.,b Department of Pharmaceutical Biotechnology, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Hesam Dorosti
- a Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences , Shiraz , Iran.,b Department of Pharmaceutical Biotechnology, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Mahboubeh Zarei
- a Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences , Shiraz , Iran.,b Department of Pharmaceutical Biotechnology, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran
| | - Younes Ghasemi
- a Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences , Shiraz , Iran.,b Department of Pharmaceutical Biotechnology, School of Pharmacy , Shiraz University of Medical Sciences , Shiraz , Iran.,c Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies , Shiraz University of Medical Sciences , Shiraz , Iran.,d Biotechnology Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
25
|
Chen HR, Lai YC, Yeh TM. Dengue virus non-structural protein 1: a pathogenic factor, therapeutic target, and vaccine candidate. J Biomed Sci 2018; 25:58. [PMID: 30037331 PMCID: PMC6057007 DOI: 10.1186/s12929-018-0462-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/13/2018] [Indexed: 02/05/2023] Open
Abstract
Dengue virus (DENV) infection is the most common mosquito-transmitted viral infection. DENV infection can cause mild dengue fever or severe dengue hemorrhagic fever (DHF)/dengue shock syndrome (DSS). Hemorrhage and vascular leakage are two characteristic symptoms of DHF/DSS. However, due to the limited understanding of dengue pathogenesis, no satisfactory therapies to treat nor vaccine to prevent dengue infection are available, and the mortality of DHF/DSS is still high. DENV nonstructural protein 1 (NS1), which can be secreted in patients’ sera, has been used as an early diagnostic marker for dengue infection for many years. However, the roles of NS1 in dengue-induced vascular leakage were described only recently. In this article, the pathogenic roles of DENV NS1 in hemorrhage and vascular leakage are reviewed, and the possibility of using NS1 as a therapeutic target and vaccine candidate is discussed.
Collapse
Affiliation(s)
- Hong-Ru Chen
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Chung Lai
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
26
|
Marzinek JK, Bag N, Huber RG, Holdbrook DA, Wohland T, Verma CS, Bond PJ. A Funneled Conformational Landscape Governs Flavivirus Fusion Peptide Interaction with Lipid Membranes. J Chem Theory Comput 2018; 14:3920-3932. [DOI: 10.1021/acs.jctc.8b00438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jan K. Marzinek
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | | | - Roland G. Huber
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | - Daniel A. Holdbrook
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | | | - Chandra S. Verma
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 63755
| | - Peter J. Bond
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| |
Collapse
|
27
|
Vanegas JM, Heinrich F, Rogers DM, Carson BD, La Bauve S, Vernon BC, Akgun B, Satija S, Zheng A, Kielian M, Rempe SB, Kent MS. Insertion of Dengue E into lipid bilayers studied by neutron reflectivity and molecular dynamics simulations. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1216-1230. [PMID: 29447917 DOI: 10.1016/j.bbamem.2018.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 02/01/2023]
Abstract
The envelope (E) protein of Dengue virus rearranges to a trimeric hairpin to mediate fusion of the viral and target membranes, which is essential for infectivity. Insertion of E into the target membrane serves to anchor E and possibly also to disrupt local order within the membrane. Both aspects are likely to be affected by the depth of insertion, orientation of the trimer with respect to the membrane normal, and the interactions that form between trimer and membrane. In the present work, we resolved the depth of insertion, the tilt angle, and the fundamental interactions for the soluble portion of Dengue E trimers (sE) associated with planar lipid bilayer membranes of various combinations of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-rac-glycerol (POPG), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), and cholesterol (CHOL) by neutron reflectivity (NR) and by molecular dynamics (MD) simulations. The results show that the tip of E containing the fusion loop (FL) is located at the interface of the headgroups and acyl chains of the outer leaflet of the lipid bilayers, in good agreement with prior predictions. The results also indicate that E tilts with respect to the membrane normal upon insertion, promoted by either the anionic lipid POPG or CHOL. The simulations show that tilting of the protein correlates with hydrogen bond formation between lysines and arginines located on the sides of the trimer close to the tip (K246, K247, and R73) and nearby lipid headgroups. These hydrogen bonds provide a major contribution to the membrane anchoring and may help to destabilize the target membrane.
Collapse
Affiliation(s)
- Juan M Vanegas
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Frank Heinrich
- National Institute of Standards and Technology Center for Neutron Research, Gaithersburg, MD, United States; Department of Physics, Carnegie Mellon University, Pittsburgh, PA, United States
| | - David M Rogers
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Bryan D Carson
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Sadie La Bauve
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Briana C Vernon
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Bulent Akgun
- National Institute of Standards and Technology Center for Neutron Research, Gaithersburg, MD, United States
| | - Sushil Satija
- National Institute of Standards and Technology Center for Neutron Research, Gaithersburg, MD, United States
| | - Aihua Zheng
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Susan B Rempe
- Sandia National Laboratories, Albuquerque, NM, United States
| | - Michael S Kent
- Sandia National Laboratories, Albuquerque, NM, United States.
| |
Collapse
|
28
|
Cecilia D, Patil J, Kakade M, Walimbe A, Alagarasu K, Anukumar B, Abraham A. Emergence of the Asian genotype of DENV-1 in South India. Virology 2017; 510:40-45. [DOI: 10.1016/j.virol.2017.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/29/2017] [Accepted: 07/03/2017] [Indexed: 10/19/2022]
|
29
|
Choudhary MC, Gupta E, Sharma S, Hasnain N, Agarwala P. Genetic signatures coupled with lineage shift characterise endemic evolution of Dengue virus serotype 2 during 2015 outbreak in Delhi, India. Trop Med Int Health 2017; 22:871-880. [DOI: 10.1111/tmi.12898] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Ekta Gupta
- Department of Virology; Institute of Liver and Biliary Sciences; New Delhi India
| | - Shvetank Sharma
- Molecular and Cellular Medicine Department; Institute of Liver and Biliary Sciences; New Delhi India
| | - Nadeem Hasnain
- Department of Virology; Institute of Liver and Biliary Sciences; New Delhi India
| | - Pragya Agarwala
- Department of Virology; Institute of Liver and Biliary Sciences; New Delhi India
| |
Collapse
|
30
|
Gangopadhyay A, Chakraborty HJ, Datta A. Targeting the dengue β-OG with serotype-specific alkaloid virtual leads. J Mol Graph Model 2017; 73:129-142. [PMID: 28279821 DOI: 10.1016/j.jmgm.2017.02.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 01/24/2017] [Accepted: 02/27/2017] [Indexed: 12/31/2022]
Abstract
The dengue envelope β-OG pocket is a crucial hinge for mediating virus-host fusion via conformational changes in the envelope to the fusion-competent form. The β-OG pocket is a small molecule target site for inhibition of virus-host fusion. As of date, the only structure of the β-OG pocket known is of serotype 2. Studies of β-OG inhibition by small molecules primarily target viral serotype 2. Envelope and β-OG sequence alignments, reveal dissimilarities across serotypes. In light of protein sequence-structure-function correlation, sequence variations suggest serotypic variations in β-OG druggability. This, together with the fact that dengue viral proteins do have serotype-specific variations of structure and function, lead to the study of the serotype-specificity of the dengue β-OG ligand binding behaviour. β-OG druggability was compared using comparative models of envelope proteins containing the β-OG pocket in four serotypes of the dengue virus. β-OG ligand binding was found to vary with respect to hydrophobicity, hydrophilicity, hydrogen bonding, van der Waals interactions with ligands and tightness of the binding site. The study also reports serotype-specific virtual leads identified from a library of 9175 alkaloids, using a consensus docking and scoring approach. The docking algorithms of Glide SP and XP, together with the Lamarckian genetic algorithm were employed for consensus docking. For consensus scoring, the Glide empirical score was employed along with the scoring function of AutoDock. A multi-dimensional lead optimisation approach was performed for optimising affinity, ligand efficiency, lipophilic ligand efficiency, ADMET and molecular torsional strains. The study proposes the serotype-specific inhibition of the β-OG for an effective inhibition of virus-host fusion, in contrast to a pan inhibitor.
Collapse
Affiliation(s)
| | - Hirak Jyoti Chakraborty
- Central Inland Fisheries Research Institute, Barrackpore, Kolkata 700120, West Bengal, India.
| | - Abhijit Datta
- Jhargram Raj College, Jhargram 721507, West Bengal, India.
| |
Collapse
|
31
|
Suwanmanee S, Luplertlop N. Dengue and Zika viruses: lessons learned from the similarities between these Aedes mosquito-vectored arboviruses. J Microbiol 2017; 55:81-89. [PMID: 28120186 DOI: 10.1007/s12275-017-6494-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 12/13/2016] [Indexed: 01/14/2023]
Abstract
The currently spreading arbovirus epidemic is having a severe impact on human health worldwide. The two most common flaviviruses, dengue virus (DENV) and Zika virus (ZIKV), are transmitted through the same viral vector, Aedes spp. mosquitoes. Since the discovery of DENV in 1943, this virus has been reported to cause around 390 million human infections per year, approximately 500,000 of which require hospitalization and over 20,000 of which are lethal. The present DENV epidemic is primarily concentrated in Southeast Asia. ZIKV, which was discovered in 1952, is another important arthropod-borne flavivirus. The neurotropic role of ZIKV has been reported in infected newborns with microcephaly and in adults with Guillain-Barre syndrome. Despite DENV and ZIKV sharing the same viral vector, their complex pathogenic natures are poorly understood, and the infections they cause do not have specific treatments or effective vaccines. Therefore, this review will describe what is currently known about the clinical characteristics, pathogenesis mechanisms, and transmission of these two viruses. Better understanding of the interrelationships between DENV and ZIKV will provide a useful perspective for developing an effective strategy for controlling both viruses in the future.
Collapse
Affiliation(s)
- San Suwanmanee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Natthanej Luplertlop
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
32
|
Protective Capacity of the Human Anamnestic Antibody Response during Acute Dengue Virus Infection. J Virol 2016; 90:11122-11131. [PMID: 27707930 PMCID: PMC5126370 DOI: 10.1128/jvi.01096-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
Half of the world's population is exposed to the risk of dengue virus infection. Although a vaccine for dengue virus is now available in a few countries, its reported overall efficacy of about 60% is not ideal. Protective immune correlates following natural dengue virus infection remain undefined, which makes it difficult to predict the efficacy of new vaccines. In this study, we address the protective capacity of dengue virus-specific antibodies that are produced by plasmablasts a few days after natural secondary infection. Among a panel of 18 dengue virus-reactive human monoclonal antibodies, four groups of antibodies were identified based on their binding properties. While antibodies targeting the fusion loop of the glycoprotein of dengue virus dominated the antibody response, two smaller groups of antibodies bound to previously undescribed epitopes in domain II of the E protein. The latter, largely serotype-cross-reactive antibodies, demonstrated increased stability of binding at pH 5. These antibodies possessed weak to moderate neutralization capacity in vitro but were the most efficacious in promoting the survival of infected mice. Our data suggest that the cross-reactive anamnestic antibody response has a protective capacity despite moderate neutralization in vitro and a moderate decrease of viremia in vivo. IMPORTANCE Antibodies can protect from symptomatic dengue virus infection. However, it is not easy to assess which classes of antibodies provide protection because in vitro assays are not always predictive of in vivo protection. During a repeat infection, dengue virus-specific immune memory cells are reactivated and large amounts of antibodies are produced. By studying antibodies cloned from patients with heterologous secondary infection, we tested the protective value of the serotype-cross-reactive “recall” or “anamnestic” response. We found that results from in vitro neutralization assays did not always correlate with the ability of the antibodies to reduce viremia in a mouse model. In addition, a decrease of viremia in mice did not necessarily improve survival. The most protective antibodies were stable at pH 5, suggesting that antibody binding in the endosomes, after the antibody-virus complex is internalized, might be important to block virus spread in the organism.
Collapse
|
33
|
Fleith RC, Lobo FP, Dos Santos PF, Rocha MM, Bordignon J, Strottmann DM, Patricio DO, Pavanelli WR, Lo Sarzi M, Santos CND, Ferguson BJ, Mansur DS. Genome-wide analyses reveal a highly conserved Dengue virus envelope peptide which is critical for virus viability and antigenic in humans. Sci Rep 2016; 6:36339. [PMID: 27805018 PMCID: PMC5090869 DOI: 10.1038/srep36339] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
Targeting regions of proteins that show a high degree of structural conservation has been proposed as a method of developing immunotherapies and vaccines that may bypass the wide genetic variability of RNA viruses. Despite several attempts, a vaccine that protects evenly against the four circulating Dengue virus (DV) serotypes remains elusive. To find critical conserved amino acids in dengue viruses, 120 complete genomes of each serotype were selected at random and used to calculate conservation scores for nucleotide and amino acid sequences. The identified peptide sequences were analysed for their structural conservation and localisation using crystallographic data. The longest, surface exposed, highly conserved peptide of Envelope protein was found to correspond to amino acid residues 250 to 270. Mutation of this peptide in DV1 was lethal, since no replication of the mutant virus was detected in human cells. Antibodies against this peptide were detected in DV naturally infected patients indicating its potential antigenicity. Hence, this study has identified a highly conserved, critical peptide in DV that is a target of antibodies in infected humans.
Collapse
Affiliation(s)
- Renata C Fleith
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Francisco P Lobo
- Department of General Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula F Dos Santos
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Mariana M Rocha
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Juliano Bordignon
- Laboratory of Molecular Virology, Instituto Carlos Chagas, Curitiba, Brazil
| | - Daisy M Strottmann
- Laboratory of Molecular Virology, Instituto Carlos Chagas, Curitiba, Brazil
| | - Daniel O Patricio
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | | | | | - Claudia N D Santos
- Laboratory of Molecular Virology, Instituto Carlos Chagas, Curitiba, Brazil
| | | | - Daniel S Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
34
|
Molecular Docking and Molecular Dynamics Simulation Studies to Predict Flavonoid Binding on the Surface of DENV2 E Protein. Interdiscip Sci 2016; 9:499-511. [PMID: 26969331 DOI: 10.1007/s12539-016-0157-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 01/22/2016] [Accepted: 02/25/2016] [Indexed: 12/26/2022]
Abstract
Dengue infections are currently estimated to be 390 million cases annually. Yet, there is no vaccine or specific therapy available. Envelope glycoprotein E (E protein) of DENV mediates viral attachment and entry into the host cells. Several flavonoids have been shown to inhibit HIV-1 and hepatitis C virus entry during the virus-host membrane fusion. In this work, molecular docking method was employed to predict the binding of nine flavonoids (baicalin, baicalein, EGCG, fisetin, glabranine, hyperoside, ladanein, quercetin and flavone) to the soluble ectodomain of DENV type 2 (DENV2) E protein. Interestingly, eight flavonoids were found to dock into the same binding pocket located between the domain I and domain II of different subunits of E protein. Consistent docking results were observed not only for the E protein structures of the DENV2-Thai and DENV2-Malaysia (a homology model) but also for the E protein structures of tick-borne encephalitis virus and Japanese encephalitis virus. In addition, molecular dynamics simulations were performed to further evaluate the interaction profile of the docked E protein-flavonoid complexes. Ile4, Gly5, Asp98, Gly100 and Val151 residues of the DENV2-My E protein that aligned to the same residues in the DENV2-Thai E protein form consistent hydrogen bond interactions with baicalein, quercetin and EGCG during the simulations. This study demonstrates flavonoids potentially form interactions with the E protein of DENV2.
Collapse
|
35
|
Marzinek JK, Lakshminarayanan R, Goh E, Huber RG, Panzade S, Verma C, Bond PJ. Characterizing the Conformational Landscape of Flavivirus Fusion Peptides via Simulation and Experiment. Sci Rep 2016; 6:19160. [PMID: 26785994 PMCID: PMC4726195 DOI: 10.1038/srep19160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/12/2015] [Indexed: 11/09/2022] Open
Abstract
Conformational changes in the envelope proteins of flaviviruses help to expose the highly conserved fusion peptide (FP), a region which is critical to membrane fusion and host cell infection, and which represents a significant target for antiviral drugs and antibodies. In principle, extended timescale atomic-resolution simulations may be used to characterize the dynamics of such peptides. However, the resultant accuracy is critically dependent upon both the underlying force field and sufficient conformational sampling. In the present study, we report a comprehensive comparison of three simulation methods and four force fields comprising a total of more than 40 μs of sampling. Additionally, we describe the conformational landscape of the FP fold across all flavivirus family members. All investigated methods sampled conformations close to available X-ray structures, but exhibited differently populated ensembles. The best force field / sampling combination was sufficiently accurate to predict that the solvated peptide fold is less ordered than in the crystallographic state, which was subsequently confirmed via circular dichroism and spectrofluorometric measurements. Finally, the conformational landscape of a mutant incapable of membrane fusion was significantly shallower than wild-type variants, suggesting that dynamics should be considered when therapeutically targeting FP epitopes.
Collapse
Affiliation(s)
- Jan K Marzinek
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543.,Bioinformatics Institute (A*STAR), 30 Biopolis Str., #07-01 Matrix, Singapore 138671
| | - Rajamani Lakshminarayanan
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 12, Singapore 169856
| | - Eunice Goh
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 12, Singapore 169856
| | - Roland G Huber
- Bioinformatics Institute (A*STAR), 30 Biopolis Str., #07-01 Matrix, Singapore 138671
| | - Sadhana Panzade
- Bioinformatics Institute (A*STAR), 30 Biopolis Str., #07-01 Matrix, Singapore 138671
| | - Chandra Verma
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543.,Bioinformatics Institute (A*STAR), 30 Biopolis Str., #07-01 Matrix, Singapore 138671.,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Peter J Bond
- National University of Singapore, Department of Biological Sciences, 14 Science Drive 4, Singapore 117543.,Bioinformatics Institute (A*STAR), 30 Biopolis Str., #07-01 Matrix, Singapore 138671
| |
Collapse
|
36
|
Chaudhury S, Ripoll DR, Wallqvist A. Structure-based pKa prediction provides a thermodynamic basis for the role of histidines in pH-induced conformational transitions in dengue virus. Biochem Biophys Rep 2015; 4:375-385. [PMID: 29124227 PMCID: PMC5669449 DOI: 10.1016/j.bbrep.2015.10.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 11/21/2022] Open
Abstract
pH-induced conformational changes in dengue virus (DENV) are critical to its ability to infect host cells. The envelope protein heterodimers that make up the viral envelope shift from a dimer to a trimer conformation at low-pH during membrane fusion. Previous studies have suggested that the ionization of histidine residues at low-pH is central to this pH-induced conformational change. We sought out to use molecular modeling with structure-based pKa prediction to provide a quantitative basis for the role of histidines in pH-induced conformational changes and identify which histidine residues were primarily responsible for this transition. We combined existing crystallographic and cryo-electron microscopy data to construct templates of the dimer and trimer conformations for the mature and immature virus. We then generated homology models for the four DENV serotypes and carried out structure-based pKa prediction using Rosetta. Our results showed that the pKa values of a subset of conserved histidines in DENV successfully capture the thermodynamics necessary to drive pH-induced conformational changes during fusion. Here, we identified the structural determinants underlying these pKa values and compare our findings with previous experimental results. Structure-based pKa prediction for histidines in dengue virus was carried out. Conserved histidine residues drive pH-dependent conformation changes. The mature dimer form of the envelop protein is destabilized at low-pH.
Collapse
Affiliation(s)
- Sidhartha Chaudhury
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Ft. Detrick, MD 21702, United States
| | - Daniel R Ripoll
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Ft. Detrick, MD 21702, United States
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Ft. Detrick, MD 21702, United States
| |
Collapse
|
37
|
Cox KS, Tang A, Chen Z, Horton MS, Yan H, Wang XM, Dubey SA, DiStefano DJ, Ettenger A, Fong RH, Doranz BJ, Casimiro DR, Vora KA. Rapid isolation of dengue-neutralizing antibodies from single cell-sorted human antigen-specific memory B-cell cultures. MAbs 2015; 8:129-40. [PMID: 26491897 PMCID: PMC4966506 DOI: 10.1080/19420862.2015.1109757] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Monitoring antigen-specific memory B cells and the antibodies they encode is important for understanding the specificity, breadth and duration of immune response to an infection or vaccination. The antibodies isolated could further help design vaccine antigens for raising relevant protective immune responses. However, developing assays to measure and isolate antigen-specific memory B cells is technically challenging due to the low frequencies of these cells that exist in the circulating blood. Here, we describe a flow cytometry method to identify and isolate dengue envelope-specific memory B cells using a labeled dengue envelope protein. We enumerated dengue-envelope specific memory B cells from a cohort of dengue seropositive donors using this direct flow cytometry assay. A more established and conventional assay, the cultured B ELISPOT, was used as a benchmark comparator. Furthermore, we were able to confirm the single-sorted memory B-cell specificity by culturing B cells and differentiating them into plasma cells using cell lines expressing CD40L. The culture supernatants were assayed for antigen binding and the ability of the antibodies to neutralize the cognate dengue virus. Moreover, we successfully isolated the heavy and light Ig sequences and expressed them as full-length recombinant antibodies to reproduce the activity seen in culture supernatants. Mapping of these antibodies revealed a novel epitope for dengue 2 virus serotype. In conclusion, we established a reproducible methodology to enumerate antigen-specific memory B cells and assay their encoded antibodies for functional characterization.
Collapse
Affiliation(s)
- Kara S Cox
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Aimin Tang
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Zhifeng Chen
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Melanie S Horton
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Hao Yan
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Xin-Min Wang
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Sheri A Dubey
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Daniel J DiStefano
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | | | | | | | - Danilo R Casimiro
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| | - Kalpit A Vora
- a Department of Infectious Diseases and Vaccines-West Point ; PA; Merck Research Laboratories; Merck & Co.; Inc. Kenilworth ; NJ ; USA
| |
Collapse
|
38
|
Chew MF, Tham HW, Rajik M, Sharifah S. Anti-dengue virus serotype 2 activity and mode of action of a novel peptide. J Appl Microbiol 2015; 119:1170-80. [DOI: 10.1111/jam.12921] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/07/2015] [Accepted: 07/21/2015] [Indexed: 11/27/2022]
Affiliation(s)
- M.-F. Chew
- Virus-Host Interaction Group; Infectious Disease Laboratory (MR3); Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Selangor Malaysia
| | - H.-W. Tham
- Virus-Host Interaction Group; Infectious Disease Laboratory (MR3); Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Selangor Malaysia
| | - M. Rajik
- Synamatix Sdn. Bhd.; Chemistry Lab 4; Enterprise 2; Technology Park Malaysia; Kuala Lumpur Malaysia
| | - S.H. Sharifah
- Virus-Host Interaction Group; Infectious Disease Laboratory (MR3); Jeffrey Cheah School of Medicine and Health Sciences; Monash University Malaysia; Selangor Malaysia
| |
Collapse
|
39
|
Mechanism of Binding to Ebola Virus Glycoprotein by the ZMapp, ZMAb, and MB-003 Cocktail Antibodies. J Virol 2015; 89:10982-92. [PMID: 26311869 DOI: 10.1128/jvi.01490-15] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/17/2015] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED Cocktails of monoclonal antibodies (MAbs) that target the surface glycoprotein (GP) of Ebola virus (EBOV) are effective in nonhuman primate models and have been used under emergency compassionate-treatment protocols in human patients. However, the amino acids that form the detailed binding epitopes for the MAbs in the ZMapp, ZMAb, and the related MB-003 cocktails have yet to be identified. Other binding properties that define how each MAb functionally interacts with GP—such as affinity, epitope conservation, and epitope accessibility—also remain largely unknown. To help define how each MAb interacts with GP, here we used comprehensive alanine-scanning mutagenesis (shotgun mutagenesis), neutralization escape, and whole virion binding to define each MAb's specific epitope, epitope accessibility, epitope conservation, and apparent affinity. Each of the six therapeutic MAbs binds nonidentical epitopes in the GP base, glycan cap, or mucin-like domain. Their apparent affinity, epitope complementarity, and epitope accessibility helps explain why MAbs 4G7 and 13C6 are more protective than 2G4 and 1H3. The mucin-like domain MAbs 6D8 and 13F6 bind with the strongest apparent affinity, helping to explain their effectiveness in vivo despite their inability to neutralize virus. IMPORTANCE Ebola virus disease (EVD) can be caused by four different filovirus family members, including Ebola virus (EBOV), which infected 10 times more people in western Africa over the last year than all previous EVD outbreaks combined, with a number of cases distributed across the globe by travelers. Cocktails of inhibitory monoclonal antibodies (MAbs), such as ZMAb, MB-003, and in particular ZMapp, have demonstrated in animal models some of the most significant therapeutic potential for treating EVD, and in 2014, 15 patients were treated with ZMapp or ZMAb under compassionate-use protocols. Here, we have defined the epitope features for the most important therapeutic MAbs against EBOV developed to date. Defining the epitopes and binding characteristics for these MAbs, as well as the commonly used reference MAb KZ52, helps explain their breadth of reactivity against different ebolavirus species, predict viral evasion against these MAbs, and design new cocktails of MAbs with improved complementarity.
Collapse
|
40
|
Abstract
DENV envelope glycoprotein (E) is responsible for interacting with host cell receptors and is the main target for the development of a dengue vaccine based on an induction of neutralizing antibodies. It is well known that DENV E glycoprotein has two potential N-linked glycosylation sites at Asn67 and Asn153. The N-glycans of E glycoprotein have been shown to influence the proper folding of the protein, its cellular localization, its interactions with receptors and its immunogenicity. However, the precise structures of the N-glycans that are attached to E glycoprotein remain elusive, although the crystal structure of DENV E has been determined. This study characterized the structures of envelope protein N-linked glycans on mature DENV-2 particles derived from insect cells via an integrated method that used both lectin microarray and MALDI-TOF-MS. By combining these methods, a high heterogeneity of DENV N-glycans was found. Five types of N-glycan were identified on DENV-2, including mannose, GalNAc, GlcNAc, fucose and sialic acid; high mannose-type N-linked oligosaccharides and the galactosylation of N-glycans were the major structures that were found. Furthermore, a complex between a glycan on DENV and the carbohydrate recognition domain (CRD) of DC-SIGN was mimicked with computational docking experiments. For the first time, this study provides a comprehensive understanding of the N-linked glycan profile of whole DENV-2 particles derived from insect cells.
Collapse
|
41
|
Context-Dependent Cleavage of the Capsid Protein by the West Nile Virus Protease Modulates the Efficiency of Virus Assembly. J Virol 2015; 89:8632-42. [PMID: 26063422 DOI: 10.1128/jvi.01253-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/02/2015] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The molecular mechanisms that define the specificity of flavivirus RNA encapsulation are poorly understood. Virions composed of the structural proteins of one flavivirus and the genomic RNA of a heterologous strain can be assembled and have been developed as live attenuated vaccine candidates for several flaviviruses. In this study, we discovered that not all combinations of flavivirus components are possible. While a West Nile virus (WNV) subgenomic RNA could readily be packaged by structural proteins of the DENV2 strain 16681, production of infectious virions with DENV2 strain New Guinea C (NGC) structural proteins was not possible, despite the very high amino acid identity between these viruses. Mutagenesis studies identified a single residue (position 101) of the DENV capsid (C) protein as the determinant for heterologous virus production. C101 is located at the P1' position of the NS2B/3 protease cleavage site at the carboxy terminus of the C protein. WNV NS2B/3 cleavage of the DENV structural polyprotein was possible when a threonine (Thr101 in strain 16681) but not a serine (Ser101 in strain NGC) occupied the P1' position, a finding not predicted by in vitro protease specificity studies. Critically, both serine and threonine were tolerated at the P1' position of WNV capsid. More extensive mutagenesis revealed the importance of flanking residues within the polyprotein in defining the cleavage specificity of the WNV protease. A more detailed understanding of the context dependence of viral protease specificity may aid the development of new protease inhibitors and provide insight into associated patterns of drug resistance. IMPORTANCE West Nile virus (WNV) and dengue virus (DENV) are mosquito-borne flaviviruses that cause considerable morbidity and mortality in humans. No specific antiflavivirus therapeutics are available for treatment of infection. Proteolytic processing of the flavivirus polyprotein is an essential step in the replication cycle and is an attractive target for antiviral development. The design of protease inhibitors has been informed by insights into the molecular details of the interactions of proteases and their substrates. In this article, studies of the processing of WNV and DENV capsid proteins by the WNV protease identified an unexpected contribution of the sequence surrounding critical residues within the cleavage site on protease specificity. This demonstration of context-dependent protease cleavage has implications for the design of chimeric flaviviruses, new therapeutics, and the interpretation of flavivirus protease substrate specificity studies.
Collapse
|
42
|
Rogers DM, Kent MS, Rempe SB. Molecular basis of endosomal-membrane association for the dengue virus envelope protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1041-52. [DOI: 10.1016/j.bbamem.2014.12.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/05/2014] [Accepted: 12/19/2014] [Indexed: 11/15/2022]
|
43
|
Tham HW, Balasubramaniam VRMT, Tejo BA, Ahmad H, Hassan SS. CPB1 of Aedes aegypti interacts with DENV2 E protein and regulates intracellular viral accumulation and release from midgut cells. Viruses 2014; 6:5028-46. [PMID: 25521592 PMCID: PMC4276941 DOI: 10.3390/v6125028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/24/2014] [Accepted: 12/11/2014] [Indexed: 11/21/2022] Open
Abstract
Aedes aegypti is a principal vector responsible for the transmission of dengue viruses (DENV). To date, vector control remains the key option for dengue disease management. To develop new vector control strategies, a more comprehensive understanding of the biological interactions between DENV and Ae. aegypti is required. In this study, a cDNA library derived from the midgut of female adult Ae. aegypti was used in yeast two-hybrid (Y2H) screenings against DENV2 envelope (E) protein. Among the many interacting proteins identified, carboxypeptidase B1 (CPB1) was selected, and its biological interaction with E protein in Ae. aegypti primary midgut cells was further validated. Our double immunofluorescent assay showed that CPB1-E interaction occurred in the endoplasmic reticulum (ER) of the Ae. aegypti primary midgut cells. Overexpression of CPB1 in mosquito cells resulted in intracellular DENV2 genomic RNA or virus particle accumulation, with a lower amount of virus release. Therefore, we postulated that in Ae. aegypti midgut cells, CPB1 binds to the E protein deposited on the ER intraluminal membranes and inhibits DENV2 RNA encapsulation, thus inhibiting budding from the ER, and may interfere with immature virus transportation to the trans-Golgi network.
Collapse
Affiliation(s)
- Hong-Wai Tham
- Virus-Host Interaction Research Group, Infectious Disease Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| | - Vinod R M T Balasubramaniam
- Virus-Host Interaction Research Group, Infectious Disease Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| | - Bimo Ario Tejo
- Department of Biotechnology and Neuroscience, Faculty of Life Science, Surya University, 15810 Tangerang, Banten, Indonesia.
| | - Hamdan Ahmad
- School of Biological Sciences, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia.
| | - Sharifah Syed Hassan
- Virus-Host Interaction Research Group, Infectious Disease Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
44
|
Exposure of epitope residues on the outer face of the chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014; 88:14364-79. [PMID: 25275138 DOI: 10.1128/jvi.01943-14] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.
Collapse
|
45
|
Davidson E, Doranz BJ. A high-throughput shotgun mutagenesis approach to mapping B-cell antibody epitopes. Immunology 2014; 143:13-20. [PMID: 24854488 DOI: 10.1111/imm.12323] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 02/06/2023] Open
Abstract
Characterizing the binding sites of monoclonal antibodies (mAbs) on protein targets, their 'epitopes', can aid in the discovery and development of new therapeutics, diagnostics and vaccines. However, the speed of epitope mapping techniques has not kept pace with the increasingly large numbers of mAbs being isolated. Obtaining detailed epitope maps for functionally relevant antibodies can be challenging, particularly for conformational epitopes on structurally complex proteins. To enable rapid epitope mapping, we developed a high-throughput strategy, shotgun mutagenesis, that enables the identification of both linear and conformational epitopes in a fraction of the time required by conventional approaches. Shotgun mutagenesis epitope mapping is based on large-scale mutagenesis and rapid cellular testing of natively folded proteins. Hundreds of mutant plasmids are individually cloned, arrayed in 384-well microplates, expressed within human cells, and tested for mAb reactivity. Residues are identified as a component of a mAb epitope if their mutation (e.g. to alanine) does not support candidate mAb binding but does support that of other conformational mAbs or allows full protein function. Shotgun mutagenesis is particularly suited for studying structurally complex proteins because targets are expressed in their native form directly within human cells. Shotgun mutagenesis has been used to delineate hundreds of epitopes on a variety of proteins, including G protein-coupled receptor and viral envelope proteins. The epitopes mapped on dengue virus prM/E represent one of the largest collections of epitope information for any viral protein, and results are being used to design better vaccines and drugs.
Collapse
|
46
|
Virus-like particle secretion and genotype-dependent immunogenicity of dengue virus serotype 2 DNA vaccine. J Virol 2014; 88:10813-30. [PMID: 25008922 DOI: 10.1128/jvi.00810-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Dengue virus (DENV), composed of four distinct serotypes, is the most important and rapidly emerging arthropod-borne pathogen and imposes substantial economic and public health burdens. We constructed candidate vaccines containing the DNA of five of the genotypes of dengue virus serotype 2 (DENV-2) and evaluated the immunogenicity, the neutralizing (Nt) activity of the elicited antibodies, and the protective efficacy elicited in mice immunized with the vaccine candidates. We observed a significant correlation between the level of in vitro virus-like particle secretion, the elicited antibody response, and the protective efficacy of the vaccines containing the DNA of the different DENV genotypes in immunized mice. However, higher total IgG antibody levels did not always translate into higher Nt antibodies against homologous and heterologous viruses. We also found that, in contrast to previous reports, more than 50% of total IgG targeted ectodomain III (EDIII) of the E protein, and a substantial fraction of this population was interdomain highly neutralizing flavivirus subgroup-cross-reactive antibodies, such as monoclonal antibody 1B7-5. In addition, the lack of a critical epitope(s) in the Sylvatic genotype virus recognized by interdomain antibodies could be the major cause of the poor protection of mice vaccinated with the Asian 1 genotype vaccine (pVD2-Asian 1) from lethal challenge with virus of the Sylvatic genotype. In conclusion, although the pVD2-Asian 1 vaccine was immunogenic, elicited sufficient titers of Nt antibodies against all DENV-2 genotypes, and provided 100% protection against challenge with virus of the homologous Asian 1 genotype and virus of the heterologous Cosmopolitan genotype, it is critical to monitor the potential emergence of Sylvatic genotype viruses, since vaccine candidates under development may not protect vaccinated humans from these viruses. IMPORTANCE Five genotype-specific dengue virus serotype 2 (DENV-2) DNA vaccine candidates were evaluated for their immunogenicity, homologous and heterologous neutralizing (Nt) antibody titers, and cross-genotype protection in a murine model. The immunity elicited by our prototype vaccine candidate (Asian 1 genotype strain 16681) in mice was protective against viruses of other genotypes but not against virus of the Sylvatic genotype, whose emergence and potential risk after introduction into the human population have previously been demonstrated. The underlying mechanism of a lack of protection elicited by the prototype vaccine may at least be contributed by the absence of a flavivirus subgroup-cross-reactive, highly neutralizing monoclonal antibody 1B7-5-like epitope in DENV-2 of the Sylvatic genotype. The DENV DNA vaccine directs the synthesis and assembly of virus-like particles (VLPs) and induces immune responses similar to those elicited by live-attenuated vaccines, and its flexibility permits the fast deployment of vaccine to combat emerging viruses, such as Sylvatic genotype viruses. The enhanced VLP secretion obtained by replacement of ectodomain I-II (EDI-II) of the Cosmopolitan genotype vaccine construct (VD2-Cosmopolitan) with the Asian 1 EDI-II elicited significantly higher total IgG and Nt antibody titers and suggests a novel approach to enhance the immunogenicity of the DNA vaccine. A DENV vaccine capable of eliciting protective immunity against viruses of existing and emerging genotypes should be the focus of future DENV vaccine development.
Collapse
|
47
|
Zheng A, Yuan F, Kleinfelter LM, Kielian M. A toggle switch controls the low pH-triggered rearrangement and maturation of the dengue virus envelope proteins. Nat Commun 2014; 5:3877. [PMID: 24846574 PMCID: PMC4063126 DOI: 10.1038/ncomms4877] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 04/14/2014] [Indexed: 11/09/2022] Open
Abstract
Immature dengue virus particles undergo a dramatic conformational change upon exposure to the acidic environment of the late secretory pathway. The interactions of the E fusion proteins and prM chaperone proteins on the virus envelope are reorganized to permit prM processing by a host protease, furin, thus priming virus for fusion and infection. Here we identify a pH-dependent toggle switch that controls this key conformational change during virus maturation. Our data show that the M region of prM interacts with E at neutral pH but is released at acidic pH, while the pr region interacts with E at acidic pH but is released at neutral pH. Alanine substitution of the conserved residue H98 in prM disrupts the switch by inhibiting dissociation of M from E at low pH, resulting in impaired prM processing and decreased virus infectivity. Thus, release of M-E interaction at low pH promotes formation of a furin-accessible intermediate.
Collapse
Affiliation(s)
- Aihua Zheng
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Fei Yuan
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Lara M Kleinfelter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
48
|
Messer WB, de Alwis R, Yount BL, Royal SR, Huynh JP, Smith SA, Crowe JE, Doranz BJ, Kahle KM, Pfaff JM, White LJ, Sariol CA, de Silva AM, Baric RS. Dengue virus envelope protein domain I/II hinge determines long-lived serotype-specific dengue immunity. Proc Natl Acad Sci U S A 2014; 111:1939-44. [PMID: 24385585 PMCID: PMC3918811 DOI: 10.1073/pnas.1317350111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The four dengue virus (DENV) serotypes, DENV-1, -2, -3, and -4, are endemic throughout tropical and subtropical regions of the world, with an estimated 390 million acute infections annually. Infection confers long-term protective immunity against the infecting serotype, but secondary infection with a different serotype carries a greater risk of potentially fatal severe dengue disease, including dengue hemorrhagic fever and dengue shock syndrome. The single most effective measure to control this threat to global health is a tetravalent DENV vaccine. To date, attempts to develop a protective vaccine have progressed slowly, partly because the targets of type-specific human neutralizing antibodies (NAbs), which are critical for long-term protection, remain poorly defined, impeding our understanding of natural immunity and hindering effective vaccine development. Here, we show that the envelope glycoprotein domain I/II hinge of DENV-3 and DENV-4 is the primary target of the long-term type-specific NAb response in humans. Transplantation of a DENV-4 hinge into a recombinant DENV-3 virus showed that the hinge determines the serotype-specific neutralizing potency of primary human and nonhuman primate DENV immune sera and that the hinge region both induces NAbs and is targeted by protective NAbs in rhesus macaques. These results suggest that the success of live dengue vaccines may depend on their ability to stimulate NAbs that target the envelope glycoprotein domain I/II hinge region. More broadly, this study shows that complex conformational antibody epitopes can be transplanted between live viruses, opening up similar possibilities for improving the breadth and specificity of vaccines for influenza, HIV, hepatitis C virus, and other clinically important viral pathogens.
Collapse
Affiliation(s)
- William B. Messer
- Department of Molecular Microbiology and Immunology and
- Division of Infectious Diseases, Department of Medicine, Oregon Health and Sciences University, Portland, OR 97239
| | - Ruklanthi de Alwis
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Boyd L. Yount
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| | - Scott R. Royal
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| | - Jeremy P. Huynh
- Department of Molecular Microbiology and Immunology and
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| | | | - James E. Crowe
- Pediatrics, and
- Pathology and
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN 37232
| | | | | | | | - Laura J. White
- Global Vaccines, Inc., Research Triangle Park, NC 27709; and
| | - Carlos A. Sariol
- Caribbean Primate Research Center and
- Departments of Microbiology and Medical Zoology and
- Internal Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico 00936
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
49
|
Fink K, Shi PY. Live attenuated vaccine: the first clinically approved dengue vaccine? Expert Rev Vaccines 2013; 13:185-8. [PMID: 24350687 DOI: 10.1586/14760584.2014.870888] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Dengue virus (DENV) is the most prevalent mosquito-borne viral pathogen in humans. There are 390 million human infections each year, with 96 million infections exhibiting disease symptoms. Currently, there is no clinically approved vaccine and antiviral for DENV. The four serotypes of DENV (DENV-1, -2, -3 and -4) have 25-40% variation at the amino acid level. Such variation has posed challenges for the development of a tetravalent vaccine and therapeutics.
Collapse
Affiliation(s)
- Katja Fink
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, Singapore
| | | |
Collapse
|