We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Targeting ‘immunogenic hotspots’ in Dengue and Zika virus: an in silico approach to a common vaccine candidate

    Dhrubajyoti Mahata‡

    School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    ‡Authors contributed equally to the paper and share first authorship.

    Search for more papers by this author

    ,
    Debangshu Mukherjee‡

    School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    ‡Authors contributed equally to the paper and share first authorship.

    Search for more papers by this author

    ,
    Kheerthana Duraivelan

    School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    ,
    Vanshika Malviya

    Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    ,
    Pratap Parida

    School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    &
    Gayatri Mukherjee

    *Author for correspondence:

    E-mail Address: gayatri.mukherjee@smst.iitkgp.ac.in

    School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India

    Published Online:https://doi.org/10.2217/fvl-2022-0104

    Aim: Dengue and Zika viruses cause significant mortality globally. Considering high sequence similarity between the viral proteins, we designed common multi-epitope vaccine candidates against these pathogens. Methods: We identified multiple T and B cell epitope-rich conserved ‘immunogenic hotspots’ from highly antigenic and phylogenetically related viral proteins and used these to design the multi-epitope vaccine (MEV) candidates, ensuring high global population coverage. Results: Four MEV candidates containing conserved immunogenic hotspots from E and NS5 proteins with the highest structural integrity could favorably interact with TLR4-MD2 complex in molecular docking studies, indicating activation of TLR-mediated immune responses. MEVs also induced memory responses in silico, hallmarks of a good vaccine candidate. Conclusion: Conserved immunogenic hotspots can be utilized to design cross-protective MEV candidates.

    Plain language summary

    Dengue and Zika viruses cause significant illness globally. Interestingly, they belong to the same family of viruses and therefore are closely related to each other. The immune response developed against one can often protect against the other or in some cases exacerbate the infection. Considering these characteristics of these viruses, we have designed a common vaccine candidate using bioinformatics-based tools, taking care to exclude the infection enhancing factors.

    Tweetable abstract

    Common multi-epitope vaccine candidates against Dengue viral strains and Zika virus have been designed in silico by utilizing multiple T and B-cell epitope-rich ‘immunogenic hotspots’, derived from the most antigenic and phylogenetically-related conserved proteins.

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1. World Health Organization. WHO report on global surveillance of epidemic-prone infectious diseases-dengue and dengue haemorrhagic fever (2001). www.who.int/csr/resources/publications/dengue/CSR_ISR_2000_1/en/ (Accessed: 5 January 2020).
    • 2. Ioos S, Mallet HP, Goffart IL, Gauthier V, Cardoso T, Herida M. Current Zika virus epidemiology and recent epidemics. Med. Mal. Infect 44, 302–307 (2014).
    • 3. Rather IA, Lone JB, Bajpai VK, Park YH. Zika virus infection during pregnancy and congenital abnormalities. Front Microbiol. 8, 581 (2017).
    • 4. Rice CM, Lenches EM, Eddy SR, Shin SJ, Sheets RL, Strauss JH. Nucleotide sequence of yellow fever virus: implications for flavivirus gene expression and evolution. Science 229, 726–733 (1985).
    • 5. Wong SS, Poon RW, Wong SC. Zika virus infection-the next wave after dengue? J. Formos Med. Assoc. 115, 226–242 (2016). •• Studies indicating Phylogenetic relatedness and cross reactive immune responses between DENV and ZIKV.
    • 6. Subramaniam KS, Lant S, Goodwin L, Grifoni A, Weiskopf D, Turtle L. Two is better than one: evidence for T-cell cross-protection between dengue and zika and implications on vaccine design. Front Immunol. 11, 517 (2020).
    • 7. Kam YW, Lee CY, Teo TH et al. Cross-reactive dengue human monoclonal antibody prevents severe pathologies and death from Zika virus infections. JCI insight 2, 8 (2017).
    • 8. Stettler K, Beltramello M, Espinosa DA et al. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection. Science 353, 823–826 (2016).
    • 9. Wen J, Shresta S. T cell immunity to Zika and dengue viral infections. J. Interferon Cytokine Res. 37, 475–479 (2017).
    • 10. Regla-Nava JA, Ngono AE, Viramontes KM et al. Cross-reactive Dengue virus-specific CD8+ T cells protect against Zika virus during pregnancy. Nat. Commun. 9, 1–14 (2018).
    • 11. Lim MQ, Kumaran EA, Tan HC et al. Cross-reactivity and anti-viral function of dengue capsid and NS3-specific memory T cells toward Zika virus. Front Immunol. 9, 2225 (2018). •• Studies about Antibody dependent Enhancement of infection in DENV and ZIKV.
    • 12. Dejnirattisai W, Supasa P, Wongwiwat W et al. Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus. Nat. Immunol. 17, 1102–1108 (2016).
    • 13. Paul LM, Carlin ER, Jenkins MM et al. Dengue virus antibodies enhance Zika virus infection. Clin. Transl. Immunology 5, e117 (2016).
    • 14. Kawiecki AB, Christofferson RC. Zika virus-induced antibody response enhances dengue virus serotype 2 replication in vitro. J. Infect. Dis. 214, 1357–1360 (2016).
    • 15. Halstead S, O'rourke E. Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody. J. Exp. Med. 146, 201–217 (1977).
    • 16. Dejnirattisai W, Jumnainsong A, Onsirisakul N et al. Cross-reacting antibodies enhance dengue virus infection in humans. Science 328, 745–748 (2010).
    • 17. Sun J, Du S, Zheng Z, Cheng G, Jin X. Defeat dengue and Zika viruses with a one-two punch of vaccine and vector blockade. Front Microbiol. 11, 362 (2020). •• Studies showing ADE caused by Dengvaxia vaccine administration.
    • 18. Halstead SB. Dengvaxia sensitizes seronegatives to vaccine enhanced disease regardless of age. Vaccine 35, 6355–6358 (2017).
    • 19. Shukla R, Ramasamy V, Shanmugam RK, Ahuja R, Khanna N. Antibody-dependent enhancement: a challenge for developing a safe dengue vaccine. Front Cell Infect. Microbiol. 10, 572681 (2020).
    • 20. Doytchinova IA, Flower DR. VaxiJen: a server for prediction of protective antigens, tumour antigens and subunit vaccines. BMC bioinformatics 8, 1–7 (2017).
    • 21. Sievers F, Wilm A, Dineen D et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).
    • 22. Sidney J, Peters B, Frahm N, Brander C, Sette A. HLA class I supertypes: a revised and updated classification. BMC Immunology 9, 1–15 (2008).
    • 23. Greenbaum J, Sidney J, Chung J, Brander C, Peters B, Sette A. Functional classification of class II human leukocyte antigen (HLA) molecules reveals seven different supertypes and a surprising degree of repertoire sharing across supertypes. Immunogenetics 63, 325–335 (2011).
    • 24. Wang P, Sidney J, Kim Y et al. Peptide binding predictions for HLA DR, DP and DQ molecules. BMC Bioinformatics 11, 1–12 (2010).
    • 25. Saha S, Raghava GPS. Prediction of continuous B-cell epitopes in an antigen using recurrent neural network. Proteins 65, 40–48 (2006).
    • 26. Kringelum JV, Lundegaard C, Lund O, Neildon M. Reliable B cell epitope predictions: impacts of method development and improved benchmarking. PLOS Comput. Biol. 8, e1002829 (2012). •• Studies showing CTxB as an peptide adjuvant in vaccine design.
    • 27. Jia S, Huang X, Li H et al. Immunogenicity evaluation of recombinant Lactobacillus casei W56 expressing bovine viral diarrhea virus E2 protein in conjunction with cholera toxin B subunit as an adjuvant. Microb. Cell Fact. 19, 1–20 (2020).
    • 28. Davod J, Fatemeh DN, Honari H, Hosseini R. Constructing and transient expression of a gene cassette containing edible vaccine elements and shigellosis, anthrax and cholera recombinant antigens in tomato. Mol. Biol. Rep. 45, 2237–2246 (2018). •• Evidence of TLR4 activation by CTxB peptide.
    • 29. Phongsisay V, Iizasa EI, Hara H, Yoshida H. Evidence for TLR4 and FcRγ-CARD9 activation by cholera toxin B subunit and its direct bindings to TREM2 and LMIR5 receptors. Mol. Immunol. 66, 463–471 (2015).
    • 30. Yang J, Anishchenko I, Park H, Peng Z, Ovchinnikov N, Baker D. Improved protein structure prediction using predicted interresidue orientations. Proc. Natl Acad Sci. USA 117, 1496–1503 (2020).
    • 31. Gasteiger E, Hoogland C, Gattiker A et al. Protein Identification and Analysis Tools on the ExPASy Server. The Proteomics Protocols Handbook, 571–607 (2005).
    • 32. Abraham MJ, Murtola T, Schulz R et al. GROMACS: high performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX 1, 19–25 (2015).
    • 33. Lindorff-Larsen K, Piana S, Palmo K et al. Improved side-chain torsion potentials for the Amber ff99SB protein force field. Proteins 78(8), 1950–1958 (2010).
    • 34. Pierce BG, Wiehe K, Hwang H, Kim BH, Vreven T, Weng Z. ZDOCK server: interactive docking prediction of protein-protein complexes and symmetric multimers. Bioinformatics 30, 1771–1773 (2014).
    • 35. Dhanda SK, Vir P, Raghava GP. Designing of interferon-gamma inducing MHC class-II binders. Biology Direct 8, 30 (2013).
    • 36. Rapin N, Lund O, Bernaschi M, Castiglione F. Computational immunology meets bioinformatics: the use of prediction tools for molecular binding in the simulation of the immune system. PLOS ONE 5(4), e9862 (2010).
    • 37. Grote A, Hiller K, Scheer M et al. JCat: a novel tool to adapt codon usage of a target gene to its potential expression host. Nucleic Acids Res. 33, 526–531 (2005).
    • 38. Valdés K, Alvarez M, Pupo M, Vázquez S, Rodríguez R, Guzmán MG. Human dengue antibodies against structural and nonstructural proteins. Clin. Diagn. Lab. Immunol. 7, 856–857 (2000).
    • 39. Lai CY, Tsai WY, Lin SR et al. Antibodies to envelope glycoprotein of dengue virus during the natural course of infection are predominantly cross-reactive and recognize epitopes containing highly conserved residues at the fusion loop of domain II. J. Virol. 82, 6631–6643 (2008).
    • 40. Dai L, Song J, Lu X et al. Structures of the Zika virus envelope protein and its complex with a flavivirus broadly protective antibody. Cell Host Microbe 19, 696–704 (2016).
    • 41. Rivino L, Kumaran EA, Jovanovic V et al. Differential targeting of viral components by CD4+ versus CD8+ T lymphocytes in dengue virus infection. J. Virol. 87, 2693–2706 (2013).
    • 42. Junjhon J, Edwards TJ, Utaipat U et al. Influence of pr-M cleavage on the heterogeneity of extracellular dengue virus particles. J. Virol. 84, 8353–8358 (2010).
    • 43. Rodenhuis-Zybert IA, Van der Schaar HM, da Silva Voorham JM et al. Immature dengue virus: a veiled pathogen? PLOS Pathog. 6, e1000718 (2010).
    • 44. Frank SA. Immunology and Evolution of Infectious Disease. Princeton University Press, NJ, USA (2020).
    • 45. Allison SL, Schalich J, Stiasny K, Mandl CW, Heinz FX. Mutational evidence for an internal fusion peptide in flavivirus envelope protein E. J. Virol. 75, 4268–4275 (2001).
    • 46. Stiasny K, Kiermayr S, Holzmann H, Heinz FX. Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites. J. Virol. 80, 9557–9568 (2006).
    • 47. Amrun SN, Yee WX, Abu Bakar F et al. Novel differential linear B-cell epitopes to identify Zika and dengue virus infections in patients. Clin. Transl. Immunology 8, e1066 (2019).
    • 48. Schymkowitz J, Borg J, Stricher F, Nys R, Rousseau F, Serrano L. The FoldX web server: an online force field. Nucleic Acids Res. 33, W382–W388 (2005).
    • 49. Buß O, Rudat J, Ochsenreither K. FoldX as protein engineering tool: better than random based approaches? Comput. Struct. Biotechnol. J. 16, 25–33 (2018). •• In silico study for vaccine design against DENV and ZIKV.
    • 50. Bhardwaj A, Sharma R, Grover A. Immuno-informatics guided designing of a multi-epitope vaccine against Dengue and Zika. J. Biomol. Struct. Dyn. 1–15 (2021).
    • 51. Shi J, Sun J, Wu M et al. Inferring protective CD8+ T cell epitopes for NS5 protein of four serotypes of Dengue Virus Chinese isolates based on HLA-A, -B and -C allelic distribution: implications for epitope-based universal vaccine design. PLOS ONE 10(9), e0138729 (2015). •• Studies reporting cross protective epitopes against ZIKV and DENV from clinical isolated and mice model.
    • 52. Wen J, Tang WW, Sheets N et al. Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8+ T cells. Nat. Microbiol. 2, 17036 (2017).
    • 53. Bashyam HS, Green S, Rothman AL. Dengue virus-reactive CD8+ T cells display quantitative and qualitative differences in their response to variant epitopes of heterologous viral serotypes. J. Immunol. 176(5), 2817–2824 (2006).
    • 54. Townsley E, Woda M, Thomas SJ et al. Distinct activation phenotype of a highly conserved novel HLA-B57-restricted epitope during dengue virus infection. Immunology 141(1), 27–38 (2014).
    • 55. Simmons CP, Dong T, Chau NV et al. Early T-cell responses to dengue virus epitopes in Vietnamese adults with secondary dengue virus infections. J. Virol. 79(9), 5665–5675 (2005).