1
|
Banico EC, Sira EMJS, Fajardo LE, Dulay ANG, Odchimar NMO, Simbulan AM, Orosco FL. Advancing one health vaccination: In silico design and evaluation of a multi-epitope subunit vaccine against Nipah virus for cross-species immunization using immunoinformatics and molecular modeling. PLoS One 2024; 19:e0310703. [PMID: 39325755 PMCID: PMC11426463 DOI: 10.1371/journal.pone.0310703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
The resurgence of the Nipah virus (NiV) in 2023 has raised concerns for another potentially severe pandemic, given its history of high mortality from previous outbreaks. Unfortunately, no therapeutics and vaccines have been available for the virus. This study used immunoinformatics and molecular modeling to design and evaluate a multi-epitope subunit vaccine targeting NiV. The designed vaccine construct aims to stimulate immune responses in humans and two other intermediate animal hosts of the virus-swine and equine. Using several epitope prediction tools, ten peptides that induced B-lymphocyte responses, 17 peptides that induced cytotoxic T-lymphocyte (CTL) responses, and 12 peptides that induced helper T-lymphocyte (HTL) responses were mapped from nine NiV protein sequences. However, the CTL and HTL-inducing peptides were reduced to ten and eight, respectively, following molecular docking and dynamics. These screened peptides exhibited stability with 30 common major histocompatibility complex (MHC) receptors found in humans, swine, and equine. All peptides were linked using peptide linkers to form the multi-epitope construct and various adjuvants were tested to enhance its immunogenicity. The vaccine construct with resuscitation-promoting factor E (RpfE) adjuvant was selected as the final design based on its favorable physicochemical properties and superior immune response profile. Molecular docking was used to visualize the interaction of the vaccine to toll-like receptor 4 (TLR4), while molecular dynamics confirmed the structural stability of this interaction. Physicochemical property evaluation and computational simulations showed that the designed vaccine construct exhibited favorable properties and elicited higher antibody titers than the six multi-epitope NiV vaccine designs available in the literature. Further in vivo and in vitro experiments are necessary to validate the immunogenicity conferred by the designed vaccine construct and its epitope components. This study demonstrates the capability of computational methodologies in rational vaccine design and highlights the potential of cross-species vaccination strategies for mitigating potential NiV threats.
Collapse
Affiliation(s)
- Edward Coralde Banico
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Ella Mae Joy Sinco Sira
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Lauren Emily Fajardo
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Albert Neil Gura Dulay
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Nyzar Mabeth Obenio Odchimar
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Fredmoore Legaspi Orosco
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
- Department of Science and Technology, S&T Fellows Program, Taguig City, Metro Manila, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila City, Metro Manila, Philippines
| |
Collapse
|
2
|
Rothen DA, Dutta SK, Krenger PS, Pardini A, Vogt ACS, Josi R, Lieknina I, Osterhaus ADME, Mohsen MO, Vogel M, Martina B, Tars K, Bachmann MF. Preclinical Development of a Novel Zika Virus-like Particle Vaccine in Combination with Tetravalent Dengue Virus-like Particle Vaccines. Vaccines (Basel) 2024; 12:1053. [PMID: 39340083 PMCID: PMC11435730 DOI: 10.3390/vaccines12091053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Declared as a Public Health Emergency in 2016 by the World Health Organization (WHO), the Zika virus (ZIKV) continues to cause outbreaks that are linked to increased neurological complications. Transmitted mainly by Aedes mosquitoes, the virus is spread mostly amongst several tropical regions with the potential of territorial expansion due to environmental and ecological changes. The ZIKV envelope protein's domain III, crucial for vaccine development due to its role in receptor binding and neutralizing antibody targeting, was integrated into sterically optimized AP205 VLPs to create an EDIII-based VLP vaccine. To increase the potential size of domains that can be accommodated by AP205, two AP205 monomers were fused into a dimer, resulting in 90 rather than 180 N-/C- termini amenable for fusion. EDIII displayed on AP205 VLPs has several immunological advantages, like a repetitive surface, a size of 20-200 nm (another PASP), and packaged bacterial RNA as adjuvants (a natural toll-like receptor 7/8 ligand). In this study, we evaluated a novel vaccine candidate for safety and immunogenicity in mice, demonstrating its ability to induce high-affinity, ZIKV-neutralizing antibodies without significant disease-enhancing properties. Due to the close genetical and structural characteristics, the same mosquito vectors, and the same ecological niche of the dengue virus and Zika virus, a vaccine covering all four Dengue viruses (DENV) serotypes as well as ZIKV would be of significant interest. We co-formulated the ZIKV vaccine with recently developed DENV vaccines based on the same AP205 VLP platform and tested the vaccine mix in a murine model. This combinatory vaccine effectively induced a strong humoral immune response and neutralized all five targeted viruses after two doses, with no significant antibody-dependent enhancement (ADE) observed. Overall, these findings highlight the potential of the AP205 VLP-based combinatory vaccine as a promising approach for providing broad protection against DENV and ZIKV infections. Further investigations and preclinical studies are required to advance this vaccine candidate toward potential use in human populations.
Collapse
Affiliation(s)
- Dominik A. Rothen
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | | | - Pascal S. Krenger
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Alessandro Pardini
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Anne-Cathrine S. Vogt
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Romano Josi
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Ilva Lieknina
- Latvian Biomedical Research & Study Centre, Ratsupites iela 1, 1067 Riga, Latvia
| | - Albert D. M. E. Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Mona O. Mohsen
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
| | - Monique Vogel
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
| | - Byron Martina
- Artemis Bio-Services, 2629 JD Delft, The Netherlands
| | - Kaspars Tars
- Latvian Biomedical Research & Study Centre, Ratsupites iela 1, 1067 Riga, Latvia
| | - Martin F. Bachmann
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, 3010 Bern, Switzerland
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
3
|
Ponne S, Kumar R, Vanmathi SM, Brilhante RSN, Kumar CR. Reverse engineering protection: A comprehensive survey of reverse vaccinology-based vaccines targeting viral pathogens. Vaccine 2024; 42:2503-2518. [PMID: 38523003 DOI: 10.1016/j.vaccine.2024.02.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024]
Abstract
Vaccines have significantly reduced the impact of numerous deadly viral infections. However, there is an increasing need to expedite vaccine development in light of the recurrent pandemics and epidemics. Also, identifying vaccines against certain viruses is challenging due to various factors, notably the inability to culture certain viruses in cell cultures and the wide-ranging diversity of MHC profiles in humans. Fortunately, reverse vaccinology (RV) efficiently overcomes these limitations and has simplified the identification of epitopes from antigenic proteins across the entire proteome, streamlining the vaccine development process. Furthermore, it enables the creation of multiepitope vaccines that can effectively account for the variations in MHC profiles within the human population. The RV approach offers numerous advantages in developing precise and effective vaccines against viral pathogens, including extensive proteome coverage, accurate epitope identification, cross-protection capabilities, and MHC compatibility. With the introduction of RV, there is a growing emphasis among researchers on creating multiepitope-based vaccines aiming to stimulate the host's immune responses against multiple serotypes, as opposed to single-component monovalent alternatives. Regardless of how promising the RV-based vaccine candidates may appear, they must undergo experimental validation to probe their protection efficacy for real-world applications. The time, effort, and resources allocated to the laborious epitope identification process can now be redirected toward validating vaccine candidates identified through the RV approach. However, to overcome failures in the RV-based approach, efforts must be made to incorporate immunological principles and consider targeting the epitope regions involved in disease pathogenesis, immune responses, and neutralizing antibody maturation. Integrating multi-omics and incorporating artificial intelligence and machine learning-based tools and techniques in RV would increase the chances of developing an effective vaccine. This review thoroughly explains the RV approach, ideal RV-based vaccine construct components, RV-based vaccines designed to combat viral pathogens, its challenges, and future perspectives.
Collapse
Affiliation(s)
- Saravanaraman Ponne
- Department of Medical Biotechnology, Aarupadai Veedu Medical College and Hospital, Vinayaka Mission's Research Foundation (Deemed to be University), Kirumampakkam, Puducherry 607402, India
| | - Rajender Kumar
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, AlbaNova University Center, Stockholm 106 91, Sweden
| | - S M Vanmathi
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to be University), Pondicherry 607402, India
| | - Raimunda Sâmia Nogueira Brilhante
- Medical Mycology Specialized Center, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Chinnadurai Raj Kumar
- Mahatma Gandhi Medical Advanced Research Institute, Sri Balaji Vidyapeeth (Deemed to be University), Pondicherry 607402, India.
| |
Collapse
|
4
|
Yasmin S, Ansari MY, Pandey K, Dikhit MR. Identification of potential vaccine targets for elicitation of host immune cells against SARS-CoV-2 by reverse vaccinology approach. Int J Biol Macromol 2024; 265:130754. [PMID: 38508555 DOI: 10.1016/j.ijbiomac.2024.130754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
The COVID-19 pandemic has emerged as a critical global health crisis, demanding urgent and effective strategies for containment. While some knowledge exists about epitope sequences recognized by human immune cells and their activation of CD8+ T cells within the HLA context, comprehensive information remains limited. This study employs reverse vaccinology to explore antigenic HLA-restricted T-cell epitopes capable of eliciting durable immunity. Screening reveals 187 consensus epitopes, with 23 offering broad population coverage worldwide, spanning over 5000 HLA alleles. Sequence alignment analysis highlights the genetic distinctiveness of these peptides from Homo sapiens and their intermediate to high TAP binding efficiency. Notably, these epitopes share 100 % sequence identity across strains from nine countries, indicating potential for a uniform protective immune response among diverse ethnic populations. Docking simulations further confirm their binding capacity with the HLA allele, validating them as promising targets for SARS-CoV-2 immune recognition. The anticipated epitopes are connected with suitable linkers and adjuvant, and then assessed for its translational efficacy within a bacterial expression vector through computational cloning. Through docking, it is observed that the chimeric vaccine construct forms lasting hydrogen bonds with Toll-like receptor (TLR4), while immune simulation illustrates an increased cytotoxic response aimed at CD8+ T cells. This comprehensive computational analysis suggests the chimeric vaccine construct's potential to provoke a robust immune response against SARS-CoV-2. By delineating these antigenic fragments, our study offers valuable insights into effective vaccine and immunotherapy development against COVID-19, contributing significantly to global efforts in combating this infectious threat.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Mohammad Yousuf Ansari
- Department of Pharmaceutical Chemistry, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India
| | - Manas Ranjan Dikhit
- Department of Bioinformatics, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India.
| |
Collapse
|
5
|
Dikhit MR, Sen A. Elucidation of conserved multi-epitope vaccine against Leishmania donovani using reverse vaccinology. J Biomol Struct Dyn 2024; 42:1293-1306. [PMID: 37054523 DOI: 10.1080/07391102.2023.2201630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/29/2023] [Indexed: 04/15/2023]
Abstract
Visceral leishmaniasis (VL) is a tropical disease that causes severe public health problems in humans when untreated. As no licensed vaccine exists against VL, we aimed to formulate a potential MHC-restricted chimeric vaccine construct against this dreadful parasitic disease. Amastin-like protein derived from L. donovani is considered to be stable, immunogenic and non-allergic. A comprehensive established framework was used to explore the set of immunogenic epitopes with estimated population coverage of 96.08% worldwide. The rigorous assessment revealed 6 promiscuous T-epitopes which can plausibly be presented by more than 66 diverse HLA alleles. Further docking and simulation study of peptide receptor complexes identified a strong and stable binding interaction with better structural compactness. The predicted epitopes were combined with appropriate linkers and adjuvant molecules and their translation efficiency was evaluated in pET28+(a), an bacterial expression vector using in-silico cloning. Molecular docking followed by MD simulation study revealed a stable interaction between chimeric vaccine construct with TLRs. Immune simulation of the chimeric vaccine constructs showed an elevated Th1 immune response against both B and T epitopes. With this, the detailed computational analysis suggested that the chimeric vaccine construct can evoke a robust immune response against Leishmania donovani infection. Future studies are required to validate the role of amastin as a promising vaccine target.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manas Ranjan Dikhit
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Abhik Sen
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| |
Collapse
|
6
|
de Sales-Neto JM, Madruga Carvalho DC, Arruda Magalhães DW, Araujo Medeiros AB, Soares MM, Rodrigues-Mascarenhas S. Zika virus: Antiviral immune response, inflammation, and cardiotonic steroids as antiviral agents. Int Immunopharmacol 2024; 127:111368. [PMID: 38103408 DOI: 10.1016/j.intimp.2023.111368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus first reported from humans in Nigeria in 1954. The first outbreak occurred in Micronesia followed by an outbreak in French Polynesia and another in Brazil when the virus was associated with numerous cases of severe neurological manifestations such as Guillain-Barre syndrome in adults and congenital zika syndrome in fetuses, particularly congenital microcephaly. Innate immunity is the first line of defense against ZIKV through triggering an antiviral immune response. Along with innate immune responses, a sufficient balance between anti- and pro-inflammatory cytokines and the amount of these cytokines are triggered to enhance the antiviral responses. Here, we reviewed the complex interplay between the mediators and signal pathways that coordinate antiviral immune response and inflammation as a key to understanding the development of the underlying diseases triggered by ZIKV. In addition, we summarize current and new therapeutic strategies for ZIKV infection, highlighting cardiotonic steroids as antiviral drugs for the development of this agent.
Collapse
Affiliation(s)
- José Marreiro de Sales-Neto
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | | | | | | | - Mariana Mendonça Soares
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Sandra Rodrigues-Mascarenhas
- Laboratory of Immunobiotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, PB, Brazil.
| |
Collapse
|
7
|
Valdes I, Gil L, Lazo L, Cobas K, Romero Y, Bruno A, Suzarte E, Pérez Y, Cabrales A, Ramos Y, Hermida L, Guillén G. Recombinant protein based on domain III and capsid regions of zika virus induces humoral and cellular immune response in immunocompetent BALB/c mice. Vaccine 2023; 41:5892-5900. [PMID: 37599141 DOI: 10.1016/j.vaccine.2023.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Zika virus infection continues to be a global concern for human health due to the high-risk association of the disease with neurological disorders and microcephaly in newborn. Nowadays, no vaccine or specific antiviral treatment is available, and the development of safe and effective vaccines is yet a challenge. In this study, we obtained a novel subunit vaccine that combines two regions of zika genome, domain III of the envelope and the capsid, in a chimeric protein in E. coli bacteria. The recombinant protein was characterized with polyclonal anti-ZIKV and anti-DENV antibodies that corroborate the specificity of the molecule. In addition, the PBMC from zika-immune donors stimulated with the ZEC recombinant antigen showed the capacity to recall the memory T cell response previously generated by the natural infection. The chimeric protein ZEC was able to self-assemble after combination with an immunomodulatory specific oligonucleotide to form aggregates. The inoculation of BALB/c mice with ZEC aggregated and not aggregated form of the protein showed a similar humoral immune response, although the aggregated variant induced more cell-mediated immunity evaluated by in vitro IFNγ secretion. In this study, we propose a novel vaccine candidate against the zika disease based on a recombinant protein that can stimulate both arms of the immune system.
Collapse
Affiliation(s)
- Iris Valdes
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba.
| | - Lázaro Gil
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Laura Lazo
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Karem Cobas
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Yaremis Romero
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Andy Bruno
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Edith Suzarte
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Yusleidi Pérez
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Ania Cabrales
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Lisset Hermida
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| | - Gerardo Guillén
- Center for Genetic Engineering and Biotechnology (CIGB), Avenue 31, Playa, P.O. Box 6162, Havana 10600, Cuba
| |
Collapse
|
8
|
Prediction of Conformational and Linear B-Cell Epitopes on Envelop Protein of Zika Virus Using Immunoinformatics Approach. Int J Pept Res Ther 2023; 29:17. [PMID: 36683612 PMCID: PMC9838338 DOI: 10.1007/s10989-022-10486-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2022] [Indexed: 01/10/2023]
Abstract
The current spread of Zika virus infection in India has become a public health issue due to the virus's possible link to birth abnormalities and neurological disorders. There is a need for enhanced vaccines or drugs as a result of its epidemic outbreak and the lack of potential medication. B-cell mediated adaptive immunity is capable of developing pathogen-specific memory that confers immunological protection. Therefore, in this study, the envelope protein of the Zika virus was retrieved from the NCBI protein database. The ABCpred and BepiPred software were used to discover linear B-cell epitopes on envelope protein. Conformational B-cell epitopes on envelope protein were identified using SEPPA 3.0 and Ellipro tools. Predicted B-cell epitopes were evaluated for allergenicity, toxicity, and antigenicity. Two consensus linear B-cell epitopes, envelope165-180 (AKVEITPNSPRAEATL) and envelope224-238 (PWHAGADTGTPHWNN) were identified using ABCpred and BepiPredtools. SEPPA 3.0 and Elliprotools predicted consensus conformational envelope98-110 (DRGWGNGCGLFGK) and envelope248-251 (AHAK) epitopes and one residue (75PRO) within envelope protein as a component of B-cell epitopes. These predicted linear and conformational B-cell epitopes will help in designing peptide vaccines that will activate the humoral response. However, in-vitro and in-vivo laboratory experimental confirmations are still needed to prove the application's feasibility.
Collapse
|
9
|
Silva AJD, Jesus ALS, Leal LRS, Silva GAS, Melo CML, Freitas AC. Pichia pastoris displaying ZIKV protein epitopes from the Envelope and NS1 induce in vitro immune activation. Vaccine 2021; 39:2545-2554. [PMID: 33814233 DOI: 10.1016/j.vaccine.2021.03.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 02/08/2023]
Abstract
The severe consequences of ZIKV infection and its emergence and re-emergence in several countries have boosted vaccines' development. Yeasts such as Pichia pastoris has been widely employed as antigen carriers for immunization against infectious agents. Components of the yeast cell wall have immunostimulatory properties, and recombinant antigens can be anchored to the cell surface to enhance the presentation to the immune system. Here we aimed at producing and anchoring ZIKV proteins in the P. pastoris surface as a vaccine approach. Expression cassettes were designed with epitopes of the Envelope and NS1 proteins. Immunofluorescence microscopy confirmed the anchoring of recombinant proteins. Yeasts' ability to stimulate immune cells was evaluated in vitro by incubation with lymphocytes and monocytes isolated from mouse spleen. P. pastoris expressing EnvNS1 epitopes promoted increased levels of IL-6, IL-10, and TNF-α cytokines and an increase in the number of CD4+, CD8+, and CD16+ lymphocytes, similarly to ZIKV. This profile is indicative of the activation of immunological cells and suggests an immunogenic potential of the proposed yeast vaccines against ZIKV, reinforcing the possibility of P. pastoris as adjuvant and carrier of antigens.
Collapse
Affiliation(s)
- Anna Jéssica D Silva
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - André Luiz S Jesus
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Lígia Rosa S Leal
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Guilherme Antonio S Silva
- Laboratório de Análises Imunológicas e Antitumorais, Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, Brazil
| | - Cristiane Moutinho L Melo
- Laboratório de Análises Imunológicas e Antitumorais, Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, Brazil
| | - Antonio C Freitas
- Laboratório de Estudos Moleculares e Terapia Experimental, Departamento de Genética, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
10
|
Evaluation of potential MHC-I allele-specific epitopes in Zika virus proteins and the effects of mutations on peptide-MHC-I interaction studied using in silico approaches. Comput Biol Chem 2021; 92:107459. [PMID: 33636637 DOI: 10.1016/j.compbiolchem.2021.107459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 11/24/2022]
Abstract
Zika virus (ZIKV) infection is a global health concern due to its association with microcephaly and neurological complications. The development of a T-cell vaccine is important to combat this disease. In this study, we propose ZIKV major histocompatibility complex I (MHC-I) epitopes based on in silico screening consensus followed by molecular docking, PRODIGY, and molecular dynamics (MD) simulation analyses. The effects of the reported mutations on peptide-MHC-I (pMHC-I) complexes were also evaluated. In general, our data indicate an allele-specific peptide-binding human leukocyte antigen (HLA) and potential epitopes. For HLA-B44, we showed that the absence of acidic residue Glu at P2, due to the loss of the electrostatic interaction with Lys45, has a negative impact on the pMHC-I complex stability and explains the low free energy estimated for the immunodominant peptide E-4 (IGVSNRDFV). Our MD data also suggest the deleterious effects of acidic residue Asp at P1 on the pMHC-I stability of HLA-B8 due to destabilization of the α-helix and β-strand. Free energy estimation further indicated that the mutation from Val to Ala at P9 of peptide E-247 (DAHAKRQTV), which was found exclusively in microcephaly samples, did not reduce HLA-B8 affinity. In contrast, the mutation from Thr to Pro at P2 of the peptide NS5-832 (VTKWTDIPY) decreased the interaction energy, number of intermolecular interactions, and adversely affected its binding mode with HLA-A1. Overall, our findings are important with regard to the design of T-cell peptide vaccines and for understanding how ZIKV escapes recognition by CD8 + T-cells.
Collapse
|
11
|
Mwaliko C, Nyaruaba R, Zhao L, Atoni E, Karungu S, Mwau M, Lavillette D, Xia H, Yuan Z. Zika virus pathogenesis and current therapeutic advances. Pathog Glob Health 2021; 115:21-39. [PMID: 33191867 PMCID: PMC7850325 DOI: 10.1080/20477724.2020.1845005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging arthropod-borne flavivirus that, upon infection, results in teratogenic effects and neurological disorders. ZIKV infections pose serious global public health concerns, prompting scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elaborated. Currently, no specific vaccines or drugs have been approved for ZIKV; however, some are undergoing clinical trials. Notably, several strategies have been used to develop antivirals, including drugs that target viral and host proteins. Additionally, drug repurposing is preferred since it is less costly and takes less time than other strategies because the drugs used have already been approved for human use. Likewise, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, peptide, protein, viral vectors, virus-like particles (VLPSs), inactivated-virus, and live-attenuated virus vaccines. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA both in vitro and in vivo. Importantly, most of these vaccines have entered clinical trials. Understanding the viral disease mechanism will provide better strategies for developing therapeutic agents against ZIKV. This review provides a comprehensive summary of the viral pathogenesis of ZIKV and current advancements in the development of vaccines and drugs against this virus.
Collapse
Affiliation(s)
- Caroline Mwaliko
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Raphael Nyaruaba
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Lu Zhao
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China
| | - Evans Atoni
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Samuel Karungu
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Matilu Mwau
- Center for Infectious and Parasitic Diseases Control Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Han Xia
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,CONTACT Han Xia ; Zhiming Yuan Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zhiming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
12
|
Rostamian M, Farasat A, Chegene Lorestani R, Nemati Zargaran F, Ghadiri K, Akya A. Immunoinformatics and molecular dynamics studies to predict T-cell-specific epitopes of four Klebsiella pneumoniae fimbriae antigens. J Biomol Struct Dyn 2020; 40:166-176. [PMID: 32820713 DOI: 10.1080/07391102.2020.1810126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Klebsiella pneumoniae (K. pneumoniae) is a causative agent of severe infections in humans. There is no publically available vaccine for K. pneumoniae infections yet. Here, using comprehensive immunoinformatics methods, T-cell-specific epitopes of four type 1 fimbriae antigens of K. pneumoniae were predicted and evaluated as potential vaccine candidates. Both CD8+ (class I) and CD4+ (class II) T-cell-specific epitopes were predicted and the epitopes similar to human proteome were excluded. Subsequently, the windows of class-II epitopes containing class-I epitopes were determined. The immunogenicity, IFN-γ production and population coverage were also estimated. Using the 3D structure of HLA and epitopes, molecular docking was carried out. Two best epitopes were selected for molecular dynamics studies. Our prediction and analyses resulted in the several dominant epitopes for each antigen. The docking results showed that all selected epitopes can bind to their restricted HLA molecules with high affinity. The molecular dynamics results indicated the stability of system with minimum possible deviation, suggesting the selected epitopes can be promising candidates for stably binding to HLA molecules. Altogether, our results suggest that the selected T-cell-specific epitopes of K. pneumoniae fimbriae antigens, particularly the two epitopes confirmed by molecular dynamics, can be applied for vaccine development. However, the in vitro and in vivo studies are required to authenticate the results of the present study.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mosayeb Rostamian
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Farasat
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Roya Chegene Lorestani
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Nemati Zargaran
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Keyghobad Ghadiri
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alisha Akya
- Infectious Diseases Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
13
|
Karwal P, Vats ID, Sinha N, Singhal A, Sehgal T, Kumari P. Therapeutic Applications of Peptides against Zika Virus: A Review. Curr Med Chem 2020; 27:3906-3923. [DOI: 10.2174/0929867326666190111115132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/22/2018] [Accepted: 12/28/2018] [Indexed: 01/27/2023]
Abstract
Zika Virus (ZIKV) belongs to the class of flavivirus that can be transmitted by Aedes
mosquitoes. The number of Zika virus caused cases of acute infections, neurological disorders and
congenital microcephaly are rapidly growing and therefore, in 2016, the World Health Organization
declared a global “Public Health Emergency of International Concern”. Anti-ZIKV therapeutic and
vaccine development strategies are growing worldwide in recent years, however, no specific and safe
treatment is available till date to save the human life. Currently, development of peptide therapeutics
against ZIKV has attracted rising attention on account of their high safety concern and low development
cost, in comparison to small therapeutic molecules and antibody-based anti-viral drugs. In present
review, an overview of ZIKV inhibition by peptide-based inhibitors including E-protein derived
peptides, antimicrobial peptides, frog skin peptides and probiotic peptides has been discussed. Peptides
inhibitors have also been reported to act against NS5, NS2B-NS3 protease and proteasome in
order to inhibit ZIKV infection. Recent advances in peptide-based therapeutics and vaccine have
been reviewed and their future promise against ZIKV infections has been explored.
Collapse
Affiliation(s)
- Preeti Karwal
- Department of Biochemistry, Deshbandhu College, University of Delhi, Delhi-110019, India
| | - Ishwar Dutt Vats
- Department of Chemistry, Deshbandhu College, University of Delhi, Delhi-110019, India
| | - Niharika Sinha
- Drug Development Laboratory Group, Gautam Buddha University, Noida, India
| | - Anchal Singhal
- Department of Chemistry, St. Joseph's College, Bengaluru, Karnataka, India
| | - Teena Sehgal
- Department of Chemistry, HMRITM, GGSIP University, New Delhi, India
| | - Pratibha Kumari
- Department of Chemistry, Deshbandhu College, University of Delhi, Delhi-110019, India
| |
Collapse
|
14
|
Abstract
With the increasing frequency of viral epidemics, vaccines to augment the human immune response system have been the medium of choice to combat viral infections. The tragic consequences of the Zika virus pandemic in South and Central America a few years ago brought the issues into sharper focus. While traditional vaccine development is time-consuming and expensive, recent advances in information technology, immunoinformatics, genetics, bioinformatics, and related sciences have opened the doors to new paradigms in vaccine design and applications.Peptide vaccines are one group of the new approaches to vaccine formulation. In this chapter, we discuss the various issues involved in the design of peptide vaccines and their advantages and shortcomings, with special reference to the Zika virus for which no drugs or vaccines are as yet available. In the process, we outline our work in this field giving a detailed step-by-step description of the protocol we follow for such vaccine design so that interested researchers can easily follow them and do their own designing. Several flowcharts and figures are included to provide a background of the software to be used and results to be anticipated.
Collapse
|
15
|
Pandey R, Dikhit MR, Kumar A, Dehury B, Pandey K, Topno RK, Das P, Bimal S. Evaluating the immunomodulatory responses of LdODC-derived MHC Class-II restricted peptides against VL. Parasite Immunol 2020; 42:e12699. [PMID: 31976563 DOI: 10.1111/pim.12699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
In a bid to develop a novel immunoprophylactic measure against visceral leishmaniasis (VL), MHC class-II-restricted epitopes of LdODC were identified by reverse vaccinology approach. Five consensus HLA-DRB1*0101-restricted epitopes were screened. The analysis revealed that the set of epitopes was presented by at least 54 diverse MHC class-II alleles. Based on in silico screening, followed by molecular dynamics simulation, population coverage analysis, and HLA cross-presentation ability, five best epitopes were evaluated. PBMCs isolated from treated VL subjects, when stimulated with synthetic peptide alone or as a cocktail of peptides, triggered a secretory IFN-γ, but not the IL-10 level. Support in this notion came from intracellular cytokine level with a considerable up-regulated IFN-γ produced by CD4+ T cells. Also, the enhanced IFN-γ seemed to be augmented with the activation of macrophages with prominent IL-12 production. Therefore, it can be concluded that the screened MHC class-II-restricted epitope hotspots derived from Leishmania ODC can trigger CD4+ T cells, which can skew macrophage functions towards protection. However, a detailed analysis can explore its potentiality as a vaccine candidate.
Collapse
Affiliation(s)
- RajKishor Pandey
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research, Hajipur, India.,Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Manas Ranjan Dikhit
- Department of Biomedical Informatics, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Avinash Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research, Hajipur, India
| | - Budheswar Dehury
- Department of Bioinformatics, ICMR-RMRC, Government of India, Bhubaneswar, India
| | - Krishna Pandey
- Departmentof Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Roshan Kamal Topno
- Departmentof Epidemiology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Sanjiva Bimal
- Division of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| |
Collapse
|
16
|
Basak SC, Majumdar S, Nandy A, Roy P, Dutta T, Vracko M, Bhattacharjee AK. Computer-Assisted and Data Driven Approaches for Surveillance, Drug Discovery, and Vaccine Design for the Zika Virus. Pharmaceuticals (Basel) 2019; 12:E157. [PMID: 31623241 PMCID: PMC6958466 DOI: 10.3390/ph12040157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Human life has been at the edge of catastrophe for millennia due diseases which emerge and reemerge at random. The recent outbreak of the Zika virus (ZIKV) is one such menace that shook the global public health community abruptly. Modern technologies, including computational tools as well as experimental approaches, need to be harnessed fast and effectively in a coordinated manner in order to properly address such challenges. In this paper, based on our earlier research, we have proposed a four-pronged approach to tackle the emerging pathogens like ZIKV: (a) Epidemiological modelling of spread mechanisms of ZIKV; (b) assessment of the public health risk of newly emerging strains of the pathogens by comparing them with existing strains/pathogens using fast computational sequence comparison methods; (c) implementation of vaccine design methods in order to produce a set of probable peptide vaccine candidates for quick synthesis/production and testing in the laboratory; and (d) designing of novel therapeutic molecules and their laboratory testing as well as validation of new drugs or repurposing of drugs for use against ZIKV. For each of these stages, we provide an extensive review of the technical challenges and current state-of-the-art. Further, we outline the future areas of research and discuss how they can work together to proactively combat ZIKV or future emerging pathogens.
Collapse
Affiliation(s)
- Subhash C Basak
- Department of Chemistry and Biochemistry, University of Minnesota, Duluth, MN 55812, USA.
| | | | - Ashesh Nandy
- Centre for Interdisciplinary Research and Education, Kolkata 700068, India.
| | - Proyasha Roy
- Centre for Interdisciplinary Research and Education, Kolkata 700068, India.
| | - Tathagata Dutta
- Centre for Interdisciplinary Research and Education, Kolkata 700068, India.
| | - Marjan Vracko
- National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia.
| | - Apurba K Bhattacharjee
- Biomedical Graduate Research Organization, Department of Microbiology and Immunology School of Medicine, Georgetown University, Washington, DC 20057, USA.
| |
Collapse
|
17
|
Prasasty VD, Grazzolie K, Rosmalena R, Yazid F, Ivan FX, Sinaga E. Peptide-Based Subunit Vaccine Design of T- and B-Cells Multi-Epitopes against Zika Virus Using Immunoinformatics Approaches. Microorganisms 2019; 7:E226. [PMID: 31370224 PMCID: PMC6722788 DOI: 10.3390/microorganisms7080226] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/15/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
The Zika virus disease, also known as Zika fever is an arboviral disease that became epidemic in the Pacific Islands and had spread to 18 territories of the Americas in 2016. Zika virus disease has been linked to several health problems such as microcephaly and the Guillain-Barré syndrome, but to date, there has been no vaccine available for Zika. Problems related to the development of a vaccine include the vaccination target, which covers pregnant women and children, and the antibody dependent enhancement (ADE), which can be caused by non-neutralizing antibodies. The peptide vaccine was chosen as a focus of this study as a safer platform to develop the Zika vaccine. In this study, a collection of Zika proteomes was used to find the best candidates for T- and B-cell epitopes using the immunoinformatics approach. The most promising T-cell epitopes were mapped using the selected human leukocyte antigen (HLA) alleles, and further molecular docking and dynamics studies showed a good peptide-HLA interaction for the best major histocompatibility complex-II (MHC-II) epitope. The most promising B-cell epitopes include four linear peptides predicted to be cross-reactive with T-cells, and conformational epitopes from two proteins accessible by antibodies in their native biological assembly. It is believed that the use of immunoinformatics methods is a promising strategy against the Zika viral infection in designing an efficacious multiepitope vaccine.
Collapse
Affiliation(s)
- Vivitri Dewi Prasasty
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta 12930, Indonesia.
| | - Karel Grazzolie
- Department of Biology, Faculty of Life Science, Surya University, Tangerang, Banten 15143, Indonesia
| | - Rosmalena Rosmalena
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Depok 16424, Indonesia
| | - Fatmawaty Yazid
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Depok 16424, Indonesia
| | - Fransiskus Xaverius Ivan
- Department of Biology, Faculty of Life Science, Surya University, Tangerang, Banten 15143, Indonesia
| | - Ernawati Sinaga
- Faculty of Biology, Universitas Nasional, Jakarta 12520, Indonesia
| |
Collapse
|
18
|
Coronado L, Rios L, Frías MT, Amarán L, Naranjo P, Percedo MI, Perera CL, Prieto F, Fonseca-Rodriguez O, Perez LJ. Positive selection pressure on E2 protein of classical swine fever virus drives variations in virulence, pathogenesis and antigenicity: Implication for epidemiological surveillance in endemic areas. Transbound Emerg Dis 2019; 66:2362-2382. [PMID: 31306567 DOI: 10.1111/tbed.13293] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/08/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022]
Abstract
Classical swine fever (CSF), caused by CSF virus (CSFV), is considered one of the most important infectious diseases with devasting consequences for the pig industry. Recent reports describe the emergence of new CSFV strains resulting from the action of positive selection pressure, due mainly to the bottleneck effect generated by ineffective vaccination. Even though a decrease in the genetic diversity of the positively selected CSFV strains has been observed by several research groups, there is little information about the effect of this selective force on the virulence degree, antigenicity and pathogenicity of this type of strains. Hence, the aim of the current study was to determine the effect of the positive selection pressure on these three parameters of CSFV strains, emerged as result of the bottleneck effects induced by improper vaccination in a CSF-endemic area. Moreover, the effect of the positively selected strains on the epidemiological surveillance system was assessed. By the combination of in vitro, in vivo and immunoinformatic approaches, we revealed that the action of the positive selection pressure induces a decrease in virulence and alteration in pathogenicity and antigenicity. However, we also noted that the evolutionary process of CSFV, especially in segregated microenvironments, could contribute to the gain-fitness event, restoring the highly virulent pattern of the circulating strains. Besides, we denoted that the presence of low virulent strains selected by bottleneck effect after inefficient vaccination can lead to a relevant challenge for the epidemiological surveillance of CSF, contributing to under-reports of the disease, favouring the perpetuation of the virus in the field. In this study, B-cell and CTL epitopes on the E2 3D-structure model were also identified. Thus, the current study provides novel and significant insights into variation in virulence, pathogenesis and antigenicity experienced by CSFV strains after the positive selection pressure effect.
Collapse
Affiliation(s)
- Liani Coronado
- Centro Nacional de Sanidad Agropecuaria (CENSA), OIE Collaborating Centre for Diagnosis and Risk Analysis of the Caribbean Region, La Habana, Cuba
| | - Liliam Rios
- Reiman Cancer Research Laboratory, Faculty of Medicine, University of New Brunswick, Saint John, New Brunswick, Canada
| | - María Teresa Frías
- Centro Nacional de Sanidad Agropecuaria (CENSA), OIE Collaborating Centre for Diagnosis and Risk Analysis of the Caribbean Region, La Habana, Cuba
| | - Laymara Amarán
- National Laboratory for Veterinary Diagnostic (NLVD), La Habana, Cuba
| | | | - María Irian Percedo
- Centro Nacional de Sanidad Agropecuaria (CENSA), OIE Collaborating Centre for Diagnosis and Risk Analysis of the Caribbean Region, La Habana, Cuba
| | - Carmen Laura Perera
- Centro Nacional de Sanidad Agropecuaria (CENSA), OIE Collaborating Centre for Diagnosis and Risk Analysis of the Caribbean Region, La Habana, Cuba
| | - Felix Prieto
- National Laboratory for Veterinary Diagnostic (NLVD), La Habana, Cuba
| | | | - Lester J Perez
- Department of Clinical Veterinary Medicine, College of Veterinary Science, University of Illinois, Urbana, IL, USA.,College of Veterinary Science, Veterinary Diagnostic Laboratory (VDL), University of Illinois, Urbana, IL, USA
| |
Collapse
|
19
|
Kumar M, Topno RK, Dikhit MR, Bhawana, Sahoo GC, Madhukar M, Pandey K, Das P. Molecular docking studies of chloroquine and its derivatives against P23 pro-zbd domain of chikungunya virus: Implication in designing of novel therapeutic strategies. J Cell Biochem 2019; 120:18298-18308. [PMID: 31310373 DOI: 10.1002/jcb.29139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 01/26/2023]
Abstract
The arthropod-transmitted chikungunya virus has emerged as an epidemic menace that causes debilitating polyarthritis. With this life-threatening impact on humans, the possible treatment requires to cure the viral infectivity. But, devoid of any vaccine against the chikungunya virus (CHIKV), there is a need to develop a novel chemotherapeutic strategy to treat this noxious infection. CHIKV carries highly compact P23pro-zbd structure that possesses potential RNA-binding surface domains which extremely influences the use of RNA template during genome replication at the time of infection and pathogenesis. Therefore, computational approaches were used to explore the novel small molecule inhibitors targeting P23pro-zbd domain. The tertiary structure was modeled and optimized using in silico approaches. The results obtained from PROCHECK (93.1% residues in favored regions), ERRAT (87.480 overall model quality) and ProSA (Z-score: -11.72) revealed the reliability of the proposed model. Interestingly, a previously reported inhibitor, chloroquine possesses good binding affinities with the target domain. In-depth analysis revealed that chloroquine derivatives such as didesethyl chloroquine hydroxyacetamide, cletoquine, hydroxychloroquine exhibited a better binding affinity. Notably, MD simulation analysis exhibited that Thr1312, Ala1355, Ala1356, Asn1357, Asp1364, Val1366, Cys1367, Ala1401, Gly1403, Ser1443, Tyr1444, Gly1445, Asn1459, and Thr1463 residues are the key amino acid responsible for stable ligand-protein interaction. The results obtained from this study provide new insights and advances the understanding to develop a new approach to consider effective and novel drug against chikungunya. However, a detailed in vivo study is required to explore its drug likeliness against this life-threatening disease.
Collapse
Affiliation(s)
- Maneesh Kumar
- Department of Virology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Roshan Kamal Topno
- Department of Epidemiology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Manas Ranjan Dikhit
- Department of Bioinformatics, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Bhawana
- Department of Virology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Ganesh Chandra Sahoo
- Department of Bioinformatics, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Major Madhukar
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India
| |
Collapse
|
20
|
Collins MH, Waggoner JJ. Detecting Vertical Zika Transmission: Emerging Diagnostic Approaches for an Emerged Flavivirus. ACS Infect Dis 2019; 5:1055-1069. [PMID: 30951637 DOI: 10.1021/acsinfecdis.9b00003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Zika virus (Zika) was recently responsible for a massive epidemic that spread throughout Latin America and beyond. Though Zika is typically asymptomatic or self-limiting, the sheer numbers of Zika infections led to the identification of unexpected phenotypes including sexual transmission, Guillain-Barré syndrome, and teratogenicity. Thousands of infants in South, Central, and North America have now been born with microcephaly or one of a number of fetal anomalies constituting the congenital Zika syndrome (CZS). Diagnosing CZS is based on a combination of clinical risk assessment and laboratory testing (which includes determining whether the mother has experienced a possible Zika infection during her pregnancy). A newborn suspected of having congenital Zika infection (due to maternal Zika infection or a birth defect described in association with congenital Zika infection) is then specifically tested for presence of Zika virus in neonatal tissue or anti-Zika IgM in the blood or cerebrospinal fluid. Though the guidelines are clear, there is room for considerable practice variation to emerge from individualized patient-provider encounters, largely due to limitations in diagnostic testing for Zika. The natural history of Zika further obscures our ability to know who, when, and how to test. Molecular diagnostics are highly specific but may not serve well those with asymptomatic infection. Serologic assays expand the diagnostic window but are complicated by cross-reactivity among related flaviviruses and passive immunity transferred from mother to baby. Furthermore, existing and emerging diagnostic tools may not be widely available due to limitations in resources and infrastructure of health systems in affected areas. Improvements in assay parameters as well as advances in platforms and deployability hold promise for optimizing diagnostic approaches for congenital Zika infection. The diagnostic tools and technologies under development must be integrated with forthcoming clinical knowledge of congenital Zika infection to fully realize the value that laboratory testing holds for diagnosing in utero mother to child transmission but also for understanding, predicting, and managing the health outcomes due to congenital Zika infection.
Collapse
Affiliation(s)
- Matthew H. Collins
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jesse J. Waggoner
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
21
|
Chesnut M, Muñoz LS, Harris G, Freeman D, Gama L, Pardo CA, Pamies D. In vitro and in silico Models to Study Mosquito-Borne Flavivirus Neuropathogenesis, Prevention, and Treatment. Front Cell Infect Microbiol 2019; 9:223. [PMID: 31338335 PMCID: PMC6629778 DOI: 10.3389/fcimb.2019.00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/11/2019] [Indexed: 01/07/2023] Open
Abstract
Mosquito-borne flaviviruses can cause disease in the nervous system, resulting in a significant burden of morbidity and mortality. Disease models are necessary to understand neuropathogenesis and identify potential therapeutics and vaccines. Non-human primates have been used extensively but present major challenges. Advances have also been made toward the development of humanized mouse models, but these models still do not fully represent human pathophysiology. Recent developments in stem cell technology and cell culture techniques have allowed the development of more physiologically relevant human cell-based models. In silico modeling has also allowed researchers to identify and predict transmission patterns and discover potential vaccine and therapeutic candidates. This review summarizes the research on in vitro and in silico models used to study three mosquito-borne flaviviruses that cause neurological disease in humans: West Nile, Dengue, and Zika. We also propose a roadmap for 21st century research on mosquito-borne flavivirus neuropathogenesis, prevention, and treatment.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Laura S. Muñoz
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Carlos A. Pardo
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
22
|
Akya A, Farasat A, Ghadiri K, Rostamian M. Identification of HLA-I restricted epitopes in six vaccine candidates of Leishmania tropica using immunoinformatics and molecular dynamics simulation approaches. INFECTION GENETICS AND EVOLUTION 2019; 75:103953. [PMID: 31284043 DOI: 10.1016/j.meegid.2019.103953] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
Abstract
In spite of numerous studies on vaccination for various species of Leishmania, research on the development of an effective vaccine for L. tropica is very scarce. In silico epitope prediction is a new way to survey the best vaccine candidates. Here, we predicted the best epitopes of six L. tropica antigens with vaccine capability against this pathogen, using highly frequent HLA-I alleles. Based on the frequent HLA alleles, the protein sequences were screened individually using four different MHC prediction applications, namely SYFPEITHI, ProPredI, BIMAS, and IEDB. Several in silico assays including clustering, human similarity exclusion, epitope conservancy prediction, investigating in experimental records, immunogenicity prediction, and prediction of population coverage were performed to narrow the results and to find the best epitopes. The selected epitopes and their restricted HLA-I alleles were docked and the final epitopes with the lowest binding energy (the highest binding affinity) were chosen. Finally, the stability and the binding properties of the best epitope-HLA-I combinations were analyzed using molecular dynamics simulation studies. We found ten potential peptides with strong binding affinity to highly frequent HLA-I alleles that can be further evaluated as vaccine targets against L. tropica.
Collapse
Affiliation(s)
- Alisha Akya
- Infectious Diseases Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Farasat
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Keyghobad Ghadiri
- Infectious Diseases Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mosayeb Rostamian
- Infectious Diseases Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
23
|
ZIKA virus entry mechanisms in human cells. INFECTION GENETICS AND EVOLUTION 2019; 69:22-29. [DOI: 10.1016/j.meegid.2019.01.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/29/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
|
24
|
Pritam M, Singh G, Swaroop S, Singh AK, Singh SP. Exploitation of reverse vaccinology and immunoinformatics as promising platform for genome-wide screening of new effective vaccine candidates against Plasmodium falciparum. BMC Bioinformatics 2019; 19:468. [PMID: 30717656 PMCID: PMC7394322 DOI: 10.1186/s12859-018-2482-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Background In the current scenario, designing of world-wide effective malaria vaccine against Plasmodium falciparum remain challenging despite the significant progress has been made in last few decades. Conventional vaccinology (isolate, inactivate and inject) approaches are time consuming, laborious and expensive; therefore, the use of computational vaccinology tools are imperative, which can facilitate the design of new and promising vaccine candidates. Results In current investigation, initially 5548 proteins of P. falciparum genome were carefully chosen for the incidence of signal peptide/ anchor using SignalP4.0 tool that resulted into 640 surface linked proteins (SLP). Out of these SLP, only 17 were predicted to contain GPI-anchors using PredGPI tool in which further 5 proteins were considered as malarial antigenic adhesins by MAAP and VaxiJen programs, respectively. In the subsequent step, T cell epitopes of 5 genome derived predicted antigenic adhesins (GDPAA) and 5 randomly selected known malarial adhesins (RSKMA) were analysed employing MHC class I and II tools of IEDB analysis resource. Finally, VaxiJen scored T cell epitopes from each antigen were considered for prediction of population coverage (PPC) analysis in the world-wide population including malaria endemic regions. The validation of the present in silico strategy was carried out by comparing the PPC of combined (MHC class I and II) predicted epitope ensemble among GDPAA (99.97%), RSKMA (99.90%) and experimentally known epitopes (EKE) of P. falciparum (97.72%) pertaining to world-wide human population. Conclusions The present study systematically screened 5 potential protective antigens from P. falciparum genome using bioinformatics tools. Interestingly, these GDPAA, RSKMA and EKE of P. falciparum epitope ensembles forecasted to contain highly promiscuous T cell epitopes, which are potentially effective for most of the world-wide human population with malaria endemic regions. Therefore, these epitope ensembles could be considered in near future for novel and significantly effective vaccine candidate against malaria. Electronic supplementary material The online version of this article (10.1186/s12859-018-2482-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manisha Pritam
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Garima Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Suchit Swaroop
- Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Akhilesh Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Satarudra Prakash Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India.
| |
Collapse
|
25
|
Salvador E, Pires de Souza G, Cotta Malaquias L, Wang T, Leomil Coelho L. Identification of relevant regions on structural and nonstructural proteins of Zika virus for vaccine and diagnostic test development: an in silico approach. New Microbes New Infect 2019; 29:100506. [PMID: 30858979 PMCID: PMC6396434 DOI: 10.1016/j.nmni.2019.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/11/2019] [Accepted: 01/15/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an arbovirus belonging to the Flaviviridae family and the genus Flavivirus. Infection with ZIKV causes a mild, self-limiting febrile illness called Zika fever. However, ZIKV infection has been recently associated with microcephaly and Guillain-Barré syndrome. Vaccines for the disease are a high priority of World Health Organization. Several studies are currently being conducted to develop a vaccine against ZIKV, but until now there is no licensed ZIKV vaccine. This study used a novel immunoinformatics approach to identify potential T-cell immunogenic epitopes present in the structural and nonstructural proteins of ZIKV. Fourteen T-cell candidate epitopes were identified on ZIKV structural and nonstructural proteins: pr36-50; C61-75; C103-117; E374-382; E477-491; NS2a90-104; NS2a174-188; NS2a179-193; NS2a190-204; NS2a195-209; NS2a200-214; NS3175-189; and NS4a82-96; NS4a99-113. Among these epitopes, only E374-382 is a human leukocyte antigen (HLA) type I restricted epitope. All identified epitopes showed a low similarity with other important flaviviruses but had a high conservation rate among the ZIKV strains and a high population coverage rate. Therefore, these predicted T-cell epitopes are potential candidates targets for development of vaccines to prevent ZIKV infection.
Collapse
Affiliation(s)
- E.A. Salvador
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - G.A. Pires de Souza
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - L.C. Cotta Malaquias
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
| | - T. Wang
- Department of Microbiology & Immunology, Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - L.F. Leomil Coelho
- Institute of Biomedical Sciences, Department of Microbiology and Immunology, Federal University of Alfenas, Minas Gerais, Brazil
- Corresponding author: L. F. Leomil Coelho, Laboratório de Vacinas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas, Rua Gabriel Monteiro, 700 Centro, Alfenasm Minas Gerais, 37130-001, Brazil.
| |
Collapse
|
26
|
Dikhit MR, Das S, Mahantesh V, Kumar A, Singh AK, Dehury B, Rout AK, Ali V, Sahoo GC, Topno RK, Pandey K, Das VNR, Bimal S, Das P. The potential HLA Class I-restricted epitopes derived from LeIF and TSA of Leishmania donovani evoke anti-leishmania CD8+ T lymphocyte response. Sci Rep 2018; 8:14175. [PMID: 30242172 PMCID: PMC6154976 DOI: 10.1038/s41598-018-32040-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
To explore new protective measure against visceral leishmaniasis, reverse vaccinology approach was employed to identify key immunogenic regions which can mediate long-term immunity. In-depth computational analysis revealed nine promiscuous epitopes which can possibly be presented by 46 human leukocyte antigen, thereby broadening the worldwide population up to 94.16%. This is of reasonable significance that most of the epitopes shared 100% sequence homology with other Leishmania species and could evoke a common pattern of protective immune response. Transporter associated with antigen processing binding affinity, molecular docking approach followed by dynamics simulation and human leukocyte antigen stabilization assay suggested that the best five optimal set of epitopes bind in between α1 and α2 binding groove with sufficient affinity and stability which allows the translocation of intact epitope to the cell surface. Fascinatingly, the human leukocyte antigen stabilization assay exhibited a modest correlation with the positive immunogenicity score predicted by class I pMHC immunogenicity predictor. A support for this notion came from ELISA and FACS analysis where the epitopes as a cocktail induced CD8+ IFN-γ and Granzyme B levels significantly in treated visceral leishmaniasis subject which suggests the immunogenic ability of the selected epitopes.
Collapse
Affiliation(s)
- Manas Ranjan Dikhit
- BioMedical Informatics Division, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India.,Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, 801507, Bihar, India
| | - Vijaya Mahantesh
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Akhilesh Kumar
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Ashish Kumar Singh
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Budheswar Dehury
- BioMedical Informatics Centre, ICMR-Regional Medical Research Centre, Bhubaneswar, 751023, Odisha, India
| | - Ajaya Kumar Rout
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, West Bengal, India
| | - Vahab Ali
- Department of Clinical Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Ganesh Chandra Sahoo
- BioMedical Informatics Division, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Roshan Kamal Topno
- Department of Epidemiology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Krishna Pandey
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - V N R Das
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Sanjiva Bimal
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India
| | - Pradeep Das
- Department of Molecular Parasitology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, 800007, Bihar, India.
| |
Collapse
|
27
|
Improved Immune Responses Against Zika Virus After Sequential Dengue and Zika Virus Infection in Humans. Viruses 2018; 10:v10090480. [PMID: 30205518 PMCID: PMC6164826 DOI: 10.3390/v10090480] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 02/02/2023] Open
Abstract
The high levels of dengue-virus (DENV) seroprevalence in areas where the Zika virus (ZIKV) is circulating and the cross-reactivity between these two viruses have raised concerns on the risk of increased ZIKV disease severity for patients with a history of previous DENV infections. To determine the role of DENV preimmunity in ZIKV infection, we analyzed the T- and B-cell responses against ZIKV in donors with or without previous DENV infection. Using peripheral blood mononuclear cells (PBMCs) from donors living in an endemic area in Colombia, we have identified, by interferon (IFN)-γ enzyme-linked immunospot (ELISPOT) assay, most of the immunodominant ZIKV T-cell epitopes in the nonstructural (NS) proteins NS1, NS3, and NS5. Analyses of the T- and B-cell responses in the same donors revealed a stronger T-cell response against peptides conserved between DENV and ZIKV, with a higher level of ZIKV-neutralizing antibodies in DENV-immune donors in comparison with DENV-naïve donors. Strikingly, the potential for antibody-mediated enhancement of ZIKV infection was reduced in donors with sequential DENV and ZIKV infection in comparison with donors with DENV infection only. Altogether, these data suggest that individuals with DENV immunity present improved immune responses against ZIKV.
Collapse
|
28
|
Kumar A, Dikhit MR, Amit A, Zaidi A, Pandey RK, Singh AK, Suman SS, Ali V, Das VNR, Pandey K, kumar V, Singh SK, Narayan S, Chourasia HK, Das P, Bimal S. Immunomodulation induced through ornithine decarboxylase DNA immunization in Balb/c mice infected with Leishmania donovani. Mol Immunol 2018; 97:33-44. [DOI: 10.1016/j.molimm.2018.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/05/2018] [Accepted: 03/07/2018] [Indexed: 12/21/2022]
|
29
|
Usman S, Naz Z, Saleem K, Bashir H, Bilal M, Sumrin A. The countermeasure for Zika virus: a hard nut to be cracked. Future Virol 2018. [DOI: 10.2217/fvl-2018-0018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The emergence of Zika virus (ZIKV) infection in America has caused a new threat worldwide, as it is linked with the increased incidence of congenital microcephaly in neonates and development of Guillain–Barré syndrome. A number of potential antiviral drugs such as sofosbuvir, BCX4430, NITD008 and 7-DMA have shown activity against ZIKV both in vitro and in vivo. Similarly, different research organizations and academic institutions are trying hard to develop a vaccine against the ZIKV. Some of these groups have received approval from the US FDA to start Phase I clinical trials. Immuno-based treatment strategies, such as use of humanized monoclonal antibodies (hE16 and CR4373) have also entered clinical trials. However, a licensed vaccine is still a long way off and efforts should be made to accelerate the evaluation procedures while minimizing the delay in licensing.
Collapse
Affiliation(s)
- Sana Usman
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| | - Zara Naz
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| | - Komal Saleem
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| | - Hamid Bashir
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| | - Muhammad Bilal
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| | - Aleena Sumrin
- Center for Applied Molecular Biology, 87-West Canal, Bank Road, University of the Punjab, Lahore-53700, Pakistan
| |
Collapse
|
30
|
Computational elucidation of novel antagonists and binding insights by structural and functional analyses of serine hydroxymethyltransferase and interaction with inhibitors. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2017.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Makhluf H, Shresta S. Development of Zika Virus Vaccines. Vaccines (Basel) 2018; 6:E7. [PMID: 29346287 PMCID: PMC5874648 DOI: 10.3390/vaccines6010007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/12/2018] [Accepted: 01/14/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that emerged as a global threat following the most recent outbreak in Brazil in 2015. ZIKV infection of pregnant women is associated with fetal abnormalities such as microcephaly, and infection of adults can lead to Guillain-Barré syndrome, an autoimmune disease characterized by neurological deficits. Although there are currently licensed vaccines for other flaviviruses, there remains an urgent need for preventative vaccines against ZIKV infection. Herein we describe the current efforts to accelerate the development of ZIKV vaccines using various platforms, including live attenuated virus, inactivated virus, DNA and RNA, viral vectors, and in silico-predicted immunogenic viral epitopes. Many of these approaches have leveraged lessons learned from past experience with Dengue and other flavivirus vaccines.
Collapse
Affiliation(s)
- Huda Makhluf
- Department of Mathematics and Natural Sciences, National University, La Jolla, CA 92037, USA.
- Center for Infectious Disease, La Jolla Institute, La Jolla, CA 92037, USA.
| | - Sujan Shresta
- Center for Infectious Disease, La Jolla Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
32
|
Dikhit MR, Kumar A, Das S, Dehury B, Rout AK, Jamal F, Sahoo GC, Topno RK, Pandey K, Das VNR, Bimal S, Das P. Identification of Potential MHC Class-II-Restricted Epitopes Derived from Leishmania donovani Antigens by Reverse Vaccinology and Evaluation of Their CD4+ T-Cell Responsiveness against Visceral Leishmaniasis. Front Immunol 2017; 8:1763. [PMID: 29312304 PMCID: PMC5735068 DOI: 10.3389/fimmu.2017.01763] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/27/2017] [Indexed: 01/09/2023] Open
Abstract
Visceral leishmaniasis (VL) is one of the most neglected tropical diseases for which no vaccine exists. In spite of extensive efforts, no successful vaccine is available against this dreadful infectious disease. To support vaccine development, an immunoinformatics approach was applied to screen potential MHC class-II-restricted epitopes that can activate the immune cells. Initially, 37 epitopes derived from six stage-dependent, overexpressed antigens were predicted, which were presented by at least 26 diverse MHC class-II allele. Based on a population coverage analysis and human leukocyte antigen cross-presentation ability, six of the 37 epitopes were selected for further analysis. Stimulation with synthetic peptide alone or as a cocktail triggered intracellular IFN-γ production. Moreover, specific IgG antibodies were detected in the serum of active VL cases against P1, P4, P5, and P6 in order to evaluate the peptide effect on the humoral immune response. Additionally, most of the peptides, except P2, were found to be non-inducers of CD4+ IL-10 against both active VL as well as treated VL subjects. This finding suggests there is no role of these peptides in the pathogenesis of Leishmania. Peptide immunogenicity was validated in BALB/c mice immunized with a cocktail of synthetic peptide emulsified in complete Freund’s adjuvant/incomplete Freund’s adjuvant. The immunized splenocytes induced strong spleen cell proliferation upon parasite re-stimulation. Furthermore, increased IFN-γ, interleukin-12, IL-17, and IL-22 production augmented with elevated nitric oxide (NO) synthesis is thought to play a crucial role in macrophage activation. In this investigation, we identified six MHC class-II-restricted epitope hotspots of Leishmania antigens that induce CD4+ Th1 and Th17 responses, which could be used to potentiate a human universal T-epitope vaccine against VL.
Collapse
Affiliation(s)
- Manas Ranjan Dikhit
- BioMedical Informatics Division, Rajendra Memorial Research Institute of Medical Sciences, Patna, India.,Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Akhilesh Kumar
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Sushmita Das
- Department of Microbiology, All India Institute of Medical Sciences, Patna, India
| | - Budheswar Dehury
- Biomedical Informatics Centre, ICMR-Regional Medical Research Centre, Odisha, India
| | - Ajaya Kumar Rout
- Biotechnology Laboratory, ICAR-Central Inland Fisheries Research Institute, Kolkata, India
| | - Fauzia Jamal
- Department of Microbiology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ganesh Chandra Sahoo
- BioMedical Informatics Division, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Roshan Kamal Topno
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Krishna Pandey
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - V N R Das
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Sanjiva Bimal
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Pradeep Das
- Department of Molecular Parasitology, Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| |
Collapse
|
33
|
Kalyanaraman N. In silico prediction of potential vaccine candidates on capsid protein of human bocavirus 1. Mol Immunol 2017; 93:193-205. [PMID: 29207326 DOI: 10.1016/j.molimm.2017.11.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/18/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Human bocavirus 1 (HBoV1) is a newly identified parvovirus that causes serious respiratory infection among children across the globe. Aim of the present study was to predict immunogenic residues located on the VP2 protein of HBoV1 towards development of epitope based vaccines. Several computational tools were employed to predict epitopes (bothT and B cell restricted) with stringent regulation for the improvement of confidence. After meticulous analysis, the peptide "TTPWTYFNFNQY" was identified as potential candidate for development of preventive vaccine. Of note, the epitope "TTPWTYFNFNQY" was found to be recognized by fifteen different alleles belonging to seven HLA supertypes (A1, A3, A24, A26, B7, B58 and B62). Further, mutational variability analysis pointed that most of the amino acids were well conserved. Docking scores obtained from ClusPro and Autodock Vina for selected epitopes displayed energetically favorable and stable interaction of peptide-HLA-I complexes. The core peptide "LLYQMPFFL" was found to recognize by wide range of HLA class II allele recognition thereby qualified as candidate for therapeutic vaccine. Five distinct linear peptides (withT cell epitope superimposition) belonging to B cells were identified in the VP2 protein. Further attention on the enlisted epitopes may shed light on the path for development of diagnostic, therapeutic and preventive tools against HBoV1 infection. Additionally, the predicted epitopes may help us to address the original antigenic sin phenomena observed during consecutive HBoV2-4 infection.
Collapse
Affiliation(s)
- Narayanan Kalyanaraman
- Viral Research Diagnostic Laboratory (VRDL), Department of Microbiology, Govt Theni Medical College, Theni, Tamil Nadu, India.
| |
Collapse
|
34
|
Sankar S, Ramamurthy M, Nandagopal B, Sridharan G. In Silico Validation of D7 Salivary Protein-derived B- and T-cell Epitopes of Aedes aegypti as Potential Vaccine to Prevent Transmission of Flaviviruses and Togaviruses to Humans. Bioinformation 2017; 13:366-375. [PMID: 29225429 PMCID: PMC5712781 DOI: 10.6026/97320630013366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/22/2017] [Indexed: 11/23/2022] Open
Abstract
Mosquito (Aedes aegyptii) salivary proteins play a crucial role in facilitating viral transmission from vector-to-host due to their role in facilitating the "blood meal" of the vector. Three main proteins, D7, aegyptin and Sialokinin play a role in this process. Using in-silico programs, we identified B- and T-cell epitopes in the mosquito salivary proteins D7 long and short form. T-cell epitopes with high affinity to the most prevalent HLA MHC class-I supertypes among different population groups was chosen. It is our postulate that these epitopes could be successful in eliciting B and T cell responses, which would decrease the vector blood meal efficiency and hence protect against host infection by certain viruses. These include causative agents like Dengue viruses, Chikungunya virus, Zika and Yellow fever viruses. These viruses are of major public health importance in several countries in the Americas, Asia and Africa. Experimental evidence exists in previously published literature showing the protective effect of antibodies to certain salivary proteins in susceptible hosts. A novel approach of immunizing humans against the vector proteins to reduce transmission of viruses is now under investigation in several laboratories. We have identified the following two B cell epitopes LAALHVTAAPLWDAKDPEQF one from D7L and the other TSEYPDRQNQIEELNKLCKN from D7S. Likewise, two T cell epitopes MTSKNELDV one from D7L and the other YILCKASAF from D7S with affinity to the predominant MHC class-I supertypes were identified towards evaluation as potential vaccine.
Collapse
Affiliation(s)
- Sathish Sankar
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore - 632055, Tamil Nadu, India
| | - Mageshbabu Ramamurthy
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore - 632055, Tamil Nadu, India
| | - Balaji Nandagopal
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore - 632055, Tamil Nadu, India
| | - Gopalan Sridharan
- Sri Sakthi Amma Institute of Biomedical Research, Sri Narayani Hospital and Research Centre, Sripuram, Vellore - 632055, Tamil Nadu, India
| |
Collapse
|
35
|
Overlapping CD8+ and CD4+ T-cell epitopes identification for the progression of epitope-based peptide vaccine from nucleocapsid and glycoprotein of emerging Rift Valley fever virus using immunoinformatics approach. INFECTION GENETICS AND EVOLUTION 2017; 56:75-91. [PMID: 29107145 PMCID: PMC7106247 DOI: 10.1016/j.meegid.2017.10.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/02/2017] [Accepted: 10/25/2017] [Indexed: 12/19/2022]
Abstract
Rift Valley fever virus (RVFV) is an emergent arthropod-borne zoonotic infectious viral pathogen which causes fatal diseases in the humans and ruminants. Currently, no effective and licensed vaccine is available for the prevention of RVFV infection in endemic as well as in non-endemic regions. So, an immunoinformatics-driven genome-wide screening approach was performed for the identification of overlapping CD8+ and CD4+ T-cell epitopes and also linear B-cell epitopes from the conserved sequences of the nucleocapsid (N) and glycoprotein (G) of RVFV. We identified overlapping 99.39% conserved 1 CD8+ T-cell epitope (MMHPSFAGM) from N protein and 100% conserved 7 epitopes (AVFALAPVV, LAVFALAPV, FALAPVVFA, VFALAPVVF, IAMTVLPAL, FFDWFSGLM, and FLLIYLGRT) from G protein and also identified IL-4 and IFN-γ induced (99.39% conserved) 1 N protein CD4+ T-cell epitope (HMMHPSFAGMVDPSL) and 100% conserved 5 G protein CD4+ T-cell epitopes (LPALAVFALAPVVFA, PALAVFALAPVVFAE, GIAMTVLPALAVFAL, GSWNFFDWFSGLMSW, and FFLLIYLGRTGLSKM). The overlapping CD8+ and CD4+ T-cell epitopes were bound with most conserved HLA-C*12:03 and HLA-DRB1*01:01, respectively with the high binding affinity (kcal/mol). The combined population coverage analysis revealed that the allele frequencies of these epitopes are high in endemic and non-endemic regions. Besides, we found 100% conserved and non-allergenic 2 decamer B-cell epitopes, GVCEVGVQAL and RVFNCIDWVH of G protein had the sequence similarity with the nonamer CD8+ T-cell epitopes, VCEVGVQAL and RVFNCIDWV, respectively. Consequently, these epitopes may be used for the development of epitope-based peptide vaccine against emerging RVFV. However, in vivo and in vitro experiments are required for their efficient use as a vaccine.
Collapse
|
36
|
María RR, Arturo CJ, Alicia JA, Paulina MG, Gerardo AO. The Impact of Bioinformatics on Vaccine Design and Development. Vaccines (Basel) 2017. [DOI: 10.5772/intechopen.69273] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
37
|
Dikhit MR, Amit A, Singh AK, Kumar A, Mansuri R, Sinha S, Topno RK, Mishra R, Das VNR, Pandey K, Sahoo GC, Ali V, Bimal S, Das P. Vaccine potential of HLA-A2 epitopes from Leishmania
Cysteine Protease Type III (CPC). Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Affiliation(s)
- M. R. Dikhit
- Department of Bioinformatics; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - A. Amit
- Department of Immunology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - A. K. Singh
- Department of Immunology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
- Department of Pathology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - A. Kumar
- Department of Immunology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - R. Mansuri
- Department of Bioinformatics; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - S. Sinha
- Department of Bioinformatics; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - R. K. Topno
- Department of Epidemiology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - R. Mishra
- Department of Clinical Medicine; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - V. N. R. Das
- Department of Clinical Medicine; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - K. Pandey
- Department of Clinical Medicine; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - G. C. Sahoo
- Department of Bioinformatics; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - V. Ali
- Department of Biochemistry; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - S. Bimal
- Department of Immunology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| | - P. Das
- Department of Molecular Parasitology; Rajendra Memorial Research Institute of Medical Sciences; Patna India
| |
Collapse
|
38
|
Identification of putative unique immunogenic ZIKV and DENV1-4 peptides for diagnostic cellular based tests. Sci Rep 2017; 7:6218. [PMID: 28740150 PMCID: PMC5524841 DOI: 10.1038/s41598-017-05980-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/14/2017] [Indexed: 11/25/2022] Open
Abstract
Since the re-emergence of Zika virus in 2014 and subsequent association with microcephaly, much work has focused on the development of a vaccine to halt its spread throughout the world. The mosquito vector that transmits this virus is widespread and responsible for the spread of other arboviridae including Dengue. Current diagnostic methods rely on serologic testing that are complicated by cross reactivity and therefore unable to distinguish Zika from Dengue infection in the absence of virus isolation. We performed an in silico analysis to identify potential epitopes that may stimulate a unique T-lymphocyte response to distinguish prior infection with Zika or Dengue. From this analysis, we not only identified epitopes unique to Zika and Dengue, but also identified epitopes unique to each Dengue serotype. These peptides contribute to a pool of peptides identified for vaccine development that can be tested in vitro to confirm immunogenicity, absence of homology and global population coverage. The current lack of accurate diagnostic testing hampers our ability to understand the scope of the epidemic, implications for vaccine implementation and complications related to monoinfection and co-infection with these two closely related viruses.
Collapse
|
39
|
Dikhit MR, Kumar A, Amit A, Dehury B, Nathsharma YP, Ansari MY, Ali V, Topno RK, Das V, Pandey K, Sahoo GC, Bimal S, Das P. Mining the Proteome of Leishmania donovani for the Development of Novel MHC Class I Restricted Epitope for the Control of Visceral Leishmaniasis. J Cell Biochem 2017; 119:378-391. [PMID: 28585770 DOI: 10.1002/jcb.26190] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/05/2017] [Indexed: 12/26/2022]
Abstract
Although, the precise host defence mechanism(s) is not completely understood, T cell-mediated immune responses is believed to play a pivotal role in controlling parasite infection. Here we target the stage dependent over expressed gene. Here, the consensus based computational approach was adopted for the screening of potential major histocompatibility complex class I restricted epitopes. Based on the computational analysis and previously published report, a set 19 antigenic proteins derived from Leishmania donovani were screened for further characterization as vaccine candidates. A total of 49 epitopes were predicted, which revealed a comprehensive binding affinity to the 40 different MHC class I supertypes. Based on the population coverage and HLA cross presentation, nine highly promiscuous epitopes such as LTYDDVWTV (P1), FLFPQRTAL(P2), FLFSNGAVV (P3), YIYNFGIRV (P4), YMTAAFAAL (P5), KLLRPFAPL (P6), FMLGWIVTI (P7), SLFERNKRV (P8), and SVWNRIFTL (P9) which have either a high or an intermediate TAP binding affinity were selected for further analysis. Theoretical population coverage analysis of polytope vaccine (P1-P9) revealed more than 92% population. Stimulation with the cocktail of peptide revealed a proliferative CD8+ T cell response and increased IFN-γ production. An upregulated NF-κB activity is thought to be play a pivotal role in T cell proliferation against the selected peptide. The Th1-type cytokine profile (presence of IFN-γ and absence of IL-10) suggests the potentiality of the cocktail of epitope as a subunit vaccine against leishmaniasis. However, the efficiency of these epitopes to trigger other Th1 cytokines and chemokines in a humanized mice model could explore its plausibility as a vaccine candidate. J. Cell. Biochem. 119: 378-391, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Manas R Dikhit
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | -
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Akhilesh Kumar
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Ajay Amit
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Budheswar Dehury
- Department of Bioinformatics, ICMR Regional Medical research Centre, Bhubaneswar, Odisha 751016, India
| | - Yangya Prasad Nathsharma
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Mohammad Yousuf Ansari
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Vahab Ali
- Departmentof Biochemistry, Rajendra Memorial Research Institute of Medical, Patna 800007, India
| | - Roshan Kamal Topno
- Department of Epidemiology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Vnr Das
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Krishna Pandey
- Department of Clinical Medicine, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Ganesh Chandra Sahoo
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Sanjiva Bimal
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| | - Pradeep Das
- Department of Bioinformatics, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India.,Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Patna 800007, India
| |
Collapse
|
40
|
Immuno-informatics based approaches to identify CD8+ T cell epitopes within the Leishmania donovani 3-ectonucleotidase in cured visceral leishmaniasis subjects. Microbes Infect 2017; 19:358-369. [DOI: 10.1016/j.micinf.2017.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 03/10/2017] [Accepted: 03/24/2017] [Indexed: 01/22/2023]
|
41
|
Jamal F, Dikhit MR, Singh MK, Shivam P, Kumari S, Pushpanjali S, Dubey AK, Kumar P, Narayan S, Gupta AK, Pandey K, Das VNR, Bimal S, Das P, Singh SK. Identification of B-cell Epitope of Leishmania donovani and its application in diagnosis of visceral leishmaniasis. J Biomol Struct Dyn 2016; 35:3569-3580. [PMID: 27892844 DOI: 10.1080/07391102.2016.1263240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Diagnosis of visceral leishmaniasis (VL) is often hindered by cross-reactions with antigens from other related parasite infections. This study aimed to develop an immunochromatographic test (ICT) which can detect the antigen present in circulating immune complexes (CICs) of VL patients using B-cell epitope-specific antibodies. MS analysis of six immunoreactive 2DE spots revealed two epitopes i.e. RFFVQGDGIGQHSLQEALERR (P1) and RRVAVLVLLDRL (P2) (From a hypothetical protein [Acc No: XP_003861458.1]). The epitope conservancy analysis suggested that the linear epitope (P1P2) is 97-100% conserved among Leishmania species and diverged from Homo sapiens (61% query coverage and 80% identity). Further, immunoinformatics analysis of hydrophilicity and flexibility confirmed the antigenicity of the peptide fragment. The linear epitope (P1P2) was synthesized (98% purity) and the purity was confirmed by high-performance liquid chromatography and MS. The indirect Enzyme linked immunosorbent assay results confirmed the presence of the corresponding antibody in VL patient's sera but not in those of healthy and other diseases. The result demonstrated a sensitivity 90%; Se Cl95% (82.16-96.27)% and specificity 100%; Sp Cl95% (84.56-100)% which indicated the possibility to be used as a diagnostic tool. Sensitivity, specificity, and diagnostic efficiency of colloidal gold conjugated anti-P1P2 antibody ICT strip was 100, 95.2, and 96.7%, respectively, which is slightly better as compared to other ICT for VL. Though, our result indicated the utility of anti-P1P2 antibody to detect CICs epitopes, a large-scale inspection in endemic and non-endemic area and in different ethnic population is needed for its validation and authentication.
Collapse
Affiliation(s)
- Fauzia Jamal
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Manas Ranjan Dikhit
- b Department of Bioinformatics , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Manish K Singh
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Pushkar Shivam
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Sarita Kumari
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Sinha Pushpanjali
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Amit K Dubey
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India.,c Department of Biotechnology , National Institute of Pharmaceutical Education and Research , Hajipur 844102 , India
| | - Prakash Kumar
- c Department of Biotechnology , National Institute of Pharmaceutical Education and Research , Hajipur 844102 , India
| | - Shyam Narayan
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Anil K Gupta
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Krishna Pandey
- d Department of Clinical Medicine , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - V N R Das
- d Department of Clinical Medicine , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Sanjiva Bimal
- e Department of Immunology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Pradeep Das
- f Department of Molecular Biology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| | - Shubhankar K Singh
- a Department of Microbiology , Rajendra Memorial Research Institute of Medical Sciences , Patna 800007 , India
| |
Collapse
|
42
|
Usman Mirza M, Rafique S, Ali A, Munir M, Ikram N, Manan A, Salo-Ahen OMH, Idrees M. Towards peptide vaccines against Zika virus: Immunoinformatics combined with molecular dynamics simulations to predict antigenic epitopes of Zika viral proteins. Sci Rep 2016; 6:37313. [PMID: 27934901 PMCID: PMC5146661 DOI: 10.1038/srep37313] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/27/2016] [Indexed: 12/16/2022] Open
Abstract
The recent outbreak of Zika virus (ZIKV) infection in Brazil has developed to a global health concern due to its likely association with birth defects (primary microcephaly) and neurological complications. Consequently, there is an urgent need to develop a vaccine to prevent or a medicine to treat the infection. In this study, immunoinformatics approach was employed to predict antigenic epitopes of Zika viral proteins to aid in development of a peptide vaccine against ZIKV. Both linear and conformational B-cell epitopes as well as cytotoxic T-lymphocyte (CTL) epitopes were predicted for ZIKV Envelope (E), NS3 and NS5 proteins. We further investigated the binding interactions of altogether 15 antigenic CTL epitopes with three class I major histocompatibility complex (MHC I) proteins after docking the peptides to the binding groove of the MHC I proteins. The stability of the resulting peptide-MHC I complexes was further studied by molecular dynamics simulations. The simulation results highlight the limits of rigid-body docking methods. Some of the antigenic epitopes predicted and analyzed in this work might present a preliminary set of peptides for future vaccine development against ZIKV.
Collapse
Affiliation(s)
- Muhammad Usman Mirza
- Center for Research in Molecular Medicine (CRiMM), The University of Lahore, Pakistan
| | - Shazia Rafique
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Amjad Ali
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Mobeen Munir
- Division of Science and Technology, University of Education Lahore, Pakistan
| | - Nazia Ikram
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Pakistan
| | - Abdul Manan
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Pakistan
| | - Outi M. H. Salo-Ahen
- Structural Bioinformatics Laboratory, Faculty of Science and Engineering, Biochemistry, Åbo Akademi University, Turku, Finland
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Pharmacy, Åbo Akademi University, Turku, Finland
| | - Muhammad Idrees
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
- Vice Chancellor Hazara University, Mansehra, Pakistan
| |
Collapse
|