Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
1.
Biotechnol Lett ; 46(3): 315-354, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38403788

ABSTRACT

The HIV-1 virus has been regarded as a catastrophe for human well-being. The global incidence of HIV-1-infected individuals is increasing. Hence, development of effective immunostimulatory molecules has recently attracted an increasing attention in the field of vaccine design against HIV-1 infection. In this study, we explored the impacts of CD40L and IFN-γ as immunostimulatory adjuvants for our candidate HIV-1 Nef vaccine in human and mouse using immunoinformatics analyses. Overall, 18 IFN-γ-based vaccine constructs (9 constructs in human and 9 constructs in mouse), and 18 CD40L-based vaccine constructs (9 constructs in human and 9 constructs in mouse) were designed. To find immunogenic epitopes, important characteristics of each component (e.g., MHC-I and MHC-II binding, and peptide-MHC-I/MHC-II molecular docking) were determined. Then, the selected epitopes were applied to create multiepitope constructs. Finally, the physicochemical properties, linear and discontinuous B cell epitopes, and molecular interaction between the 3D structure of each construct and CD40, IFN-γ receptor or toll-like receptors (TLRs) were predicted. Our data showed that the full-length CD40L and IFN-γ linked to the N-terminal region of Nef were capable of inducing more effective immune response than multiepitope vaccine constructs. Moreover, molecular docking of the non-allergenic full-length- and epitope-based CD40L and IFN-γ constructs to their cognate receptors, CD40 and IFN-γ receptors, and TLRs 4 and 5 in mouse were more potent than in human. Generally, these findings suggest that the full forms of these adjuvants could be more efficient for improvement of HIV-1 Nef vaccine candidate compared to the designed multiepitope-based constructs.


Subject(s)
AIDS Vaccines , HIV-1 , Interferon-gamma , Vaccines, Subunit , nef Gene Products, Human Immunodeficiency Virus , HIV-1/immunology , Animals , Vaccines, Subunit/immunology , Vaccines, Subunit/chemistry , Mice , AIDS Vaccines/immunology , AIDS Vaccines/chemistry , Humans , Interferon-gamma/metabolism , Interferon-gamma/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , nef Gene Products, Human Immunodeficiency Virus/chemistry , Adjuvants, Immunologic/pharmacology , Molecular Docking Simulation , HIV Infections/prevention & control , HIV Infections/immunology , HIV Infections/virology , CD40 Ligand/immunology , CD40 Ligand/chemistry , Computer Simulation , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes/immunology , Epitopes/chemistry , Protein Subunit Vaccines
2.
Comput Biol Med ; 170: 108056, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301512

ABSTRACT

The Nipah virus (NPV) is a highly lethal virus, known for its significant fatality rate. The virus initially originated in Malaysia in 1998 and later led to outbreaks in nearby countries such as Bangladesh, Singapore, and India. Currently, there are no specific vaccines available for this virus. The current work employed the reverse vaccinology method to conduct a comprehensive analysis of the entire proteome of the NPV virus. The aim was to identify and choose the most promising antigenic proteins that could serve as potential candidates for vaccine development. We have also designed B and T cell epitopes-based vaccine candidate using immunoinformatics approach. We have identified a total of 5 novel Cytotoxic T Lymphocytes (CTL), 5 Helper T Lymphocytes (HTL), and 6 linear B-cell potential antigenic epitopes which are novel and can be used for further vaccine development against Nipah virus. Then we performed the physicochemical properties, antigenic, immunogenic and allergenicity prediction of the designed vaccine candidate against NPV. Further, Computational analysis indicated that these epitopes possessed highly antigenic properties and were capable of interacting with immune receptors. The designed vaccine were then docked with the human immune receptors, namely TLR-2 and TLR-4 showed robust interaction with the immune receptor. Molecular dynamics simulations demonstrated robust binding and good dynamics. After numerous dosages at varied intervals, computational immune response modeling showed that the immunogenic construct might elicit a significant immune response. In conclusion, the immunogenic construct shows promise in providing protection against NPV, However, further experimental validation is required before moving to clinical trials.


Subject(s)
Nipah Virus , Humans , Immunoinformatics , Vaccines, Subunit/chemistry , Epitopes, B-Lymphocyte/chemistry , Molecular Dynamics Simulation , Vaccine Development , Computational Biology/methods , Molecular Docking Simulation
3.
BMC Immunol ; 24(1): 46, 2023 11 18.
Article in English | MEDLINE | ID: mdl-37980458

ABSTRACT

Multi-epitope polypeptide vaccines, a fusion protein, often have a string-of-beads system composed of various specific peptide epitopes, potential adjuvants, and linkers. When choosing the sequence of various segments and linkers, many alternatives are available. These variables can influence the vaccine's effectiveness through their effects on physicochemical properties and polypeptide tertiary structure.The most conserved antigens were discovered using BLASTn. To forecast the proteins' subcellular distribution, PSORTb 3.0.2 was used. Vaxign was used for the preliminary screening and antigenicity assessment. Protein solubility was also predicted using the ccSOL omics. Using PRED-TMBB, it was anticipated that the protein would localize across membranes. The IEDB and BepiPred-2.0 databases were used to predict the immunogenicity of B cell epitopes. A multi-epitope construct was developed and analyzed to evaluate. Twenty epitopes from A. baumannii's outer membrane protein (omp) were included in the vaccination. TLR4 agonist explosibility was investigated. The physicochemical characteristics, secondary and tertiary structures, and B-cell epitopes of vaccine constructs were assessed. Additionally, docking and MD experiments were used to examine the relationship between TLR4 and its agonist.Thirteen antigens were discovered, and eight of the 13 chosen proteins were predicted to be surface proteins. The 34 kDa outer membrane protein, Omp38, Omp W, CarO, putative porin, OmpA, were chosen as having the right antigenicity (≥0.5). FhuE and CdiA were eliminated from further study because of their low antigenicity. The vaccine design was developed by combining the most effective 10 B-cell and 10 MHC-I/MHCII combined coverage epitopes. The molecular formula of the vaccine was determined to be C1718H2615N507O630S17. The vaccine form has a molecular weight of 40,996.70 Da and 47 negatively charged residues (Asp + Glu), whereas 28 positively charged residues (Arg + Lys). The estimated half-life was 7.2 hours (mammalian reticulocytes, in vitro), > 20 hours (yeast, in vivo) and > 10 hours (Escherichia coli, in vivo) for the vaccine. The multi-epitope vaccine insertion is carried via the expression vector pcDNA3.1 (+).The multi-epitope vaccine may stimulate humoral and cellular immune responses, according to our findings, and it may be a candidate for an A. baumannii vaccine.


Subject(s)
Acinetobacter baumannii , Vaccines, DNA , Animals , Toll-Like Receptor 4 , Epitopes, B-Lymphocyte , Peptides , Membrane Proteins , Epitopes, T-Lymphocyte , Computational Biology , Molecular Docking Simulation , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Mammals
4.
Altern Lab Anim ; 51(5): 313-322, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37548284

ABSTRACT

The constant evolution of pathogenic viral variants and the emergence of new viruses have reinforced the need for broad-spectrum vaccines to combat such threats. The spread of new viral variants leading to epidemic and pandemic infection can be effectively contained, if broad-spectrum vaccines effective against the newer viral variants are readily available. The development of broad-spectrum, pan-neutralising antibodies against viruses which, in general terms, are very antigenically different - such as HIV, influenza virus and paramyxoviruses - has been reported in the literature. The amino acid sequences used to generate a range of approved recombinant anti-viral vaccines were analysed by using in silico methods, with the aim of identifying highly antigenic peptide regions that may be suitable for the development of broad-spectrum peptide-based anti-viral vaccines. This was achieved through the use of open-source data, an algorithm-driven probability matrix, and published in silico prediction tools (SVMTriP, IEDB-AR, VaxiJen 2.0, AllergenFP v. 1.0, AllerTOP v. 2.0, ToxinPred and ProtParam) to evaluate antigenicity, MHC-I and MHC-II binding potential, immunogenicity, allergenicity, toxicity and physicochemical properties. We report a pan-antigenic peptide region with strong affinity for MHC-I and MHC-II, and good immunogenic potential. According to the output from the relevant in silico tools, the peptide was predicted to be non-toxic, non-allergic and to possess the desired physicochemical properties for potentially successful vaccine production. With further investigation and optimisation, this peptide could be considered for use in the development of a broad-spectrum anti-viral vaccine that may protect against emerging new viruses. Our approach of using in silico methods to identify candidate antigenic peptides with the desired physicochemical properties could potentially circumvent the use of some animal studies for peptide vaccine candidate evaluation.


Subject(s)
Influenza Vaccines , Orthomyxoviridae , Animals , Peptides , Amino Acid Sequence , Vaccines, Synthetic , Vaccines, Subunit/chemistry
5.
Comput Biol Med ; 163: 107233, 2023 09.
Article in English | MEDLINE | ID: mdl-37422941

ABSTRACT

In the recent past several vaccines were developed to combat the COVID-19 disease. Unfortunately, the protective efficacy of the current vaccines has been reduced due to the high mutation rate in SARS-CoV-2. Here, we successfully implemented a coevolution based immunoinformatics approach to design an epitope-based peptide vaccine considering variability in spike protein of SARS-CoV-2. The spike glycoprotein was investigated for B- and T-cell epitope prediction. Identified T-cell epitopes were mapped on previously reported coevolving amino acids in the spike protein to introduce mutation. The non-mutated and mutated vaccine components were constructed by selecting epitopes showing overlapping with the predicted B-cell epitopes and highest antigenicity. Selected epitopes were linked with the help of a linker to construct a single vaccine component. Non-mutated and mutated vaccine component sequences were modelled and validated. The in-silico expression level of the vaccine constructs (non-mutated and mutated) in E. coli K12 shows promising results. The molecular docking analysis of vaccine components with toll-like receptor 5 (TLR5) demonstrated strong binding affinity. The time series calculations including root mean square deviation (RMSD), radius of gyration (RGYR), and energy of the system over 100 ns trajectory obtained from all atom molecular dynamics simulation showed stability of the system. The combined coevolutionary and immunoinformatics approach used in this study will certainly help to design an effective peptide vaccine that may work against different strains of SARS-CoV-2. Moreover, the strategy used in this study can be implemented on other pathogens.


Subject(s)
COVID-19 , Viral Vaccines , Humans , SARS-CoV-2 , COVID-19/prevention & control , Molecular Docking Simulation , COVID-19 Vaccines , Spike Glycoprotein, Coronavirus/chemistry , Escherichia coli , Viral Vaccines/chemistry , Epitopes, T-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Computational Biology/methods
6.
BMC Bioinformatics ; 24(1): 231, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37271819

ABSTRACT

When it was first introduced in 2000, reverse vaccinology was defined as an in silico approach that begins with the pathogen's genomic sequence. It concludes with a list of potential proteins with a possible, but not necessarily, list of peptide candidates that need to be experimentally confirmed for vaccine production. During the subsequent years, reverse vaccinology has dramatically changed: now it consists of a large number of bioinformatics tools and processes, namely subtractive proteomics, computational vaccinology, immunoinformatics, and in silico related procedures. However, the state of the art of reverse vaccinology still misses the ability to predict the efficacy of the proposed vaccine formulation. Here, we describe how to fill the gap by introducing an advanced immune system simulator that tests the efficacy of a vaccine formulation against the disease for which it has been designed. As a working example, we entirely apply this advanced reverse vaccinology approach to design and predict the efficacy of a potential vaccine formulation against influenza H5N1. Climate change and melting glaciers are critical due to reactivating frozen viruses and emerging new pandemics. H5N1 is one of the potential strains present in icy lakes that can raise a pandemic. Investigating structural antigen protein is the most profitable therapeutic pipeline to generate an effective vaccine against H5N1. In particular, we designed a multi-epitope vaccine based on predicted epitopes of hemagglutinin and neuraminidase proteins that potentially trigger B-cells, CD4, and CD8 T-cell immune responses. Antigenicity and toxicity of all predicted CTL, Helper T-lymphocytes, and B-cells epitopes were evaluated, and both antigenic and non-allergenic epitopes were selected. From the perspective of advanced reverse vaccinology, the Universal Immune System Simulator, an in silico trial computational framework, was applied to estimate vaccine efficacy using a cohort of 100 digital patients.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza, Human , Humans , Influenza, Human/prevention & control , Vaccinology/methods , Vaccine Efficacy , Epitopes, B-Lymphocyte , Proteins , Computational Biology/methods , Immune System , Epitopes, T-Lymphocyte/chemistry , Molecular Docking Simulation , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 39(6): 494-500, 2023 Jun.
Article in Chinese | MEDLINE | ID: mdl-37340917

ABSTRACT

Objectives To develop a multi-stage and multi-epitope vaccine, which consists of epitopes from the early secretory and latency-associated antigens of Mycobacterium tuberculosis (MTB). Methods The B-cell, cytotoxic T-lymphocyte (CTL) and helper T-lymphocyte (HTL) epitopes of 12 proteins were predicted using an immunoinformatics. The epitopes with antigenicity, without cytotoxicity and sensitization, were further screened to construct the multi-epitope vaccine. Furthermore, the proposed vaccine underwent physicochemical properties analysis and secondary structure prediction as well as 3D structure modeling, refinement and validation. Then the refined model was docked with TLR4. Finally, an immune simulation of the vaccine was carried out. Results The proposed vaccine, which consists of 12 B-cell, 11 CTL and 12 HTL epitopes, had a flexible and stable globular conformation as well as a thermostable and hydrophilic structure. A stable interaction of the vaccine with TLR4 was confirmed by molecular docking. The efficiency of the candidate vaccine to trigger effective cellular and humoral immune responses was assessed by immune simulation. Conclusion A multi-stage multi-epitope MTB vaccine construction strategy based on immunoinformatics is proposed, which is expected to prevent both active and latent MTB infection.


Subject(s)
Mycobacterium tuberculosis , Mycobacterium tuberculosis/metabolism , Molecular Docking Simulation , Toll-Like Receptor 4 , Epitopes, T-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Computational Biology/methods
8.
Curr Pharm Des ; 29(19): 1504-1515, 2023.
Article in English | MEDLINE | ID: mdl-37073655

ABSTRACT

BACKGROUND: Borrelia burgdorferi is regarded as an extremely dangerous bacteria causing infectious disease in humans, resulting in musculoskeletal pain, fatigue, fever and cardiac symptom. Because of all alarming concerns, no such prophylaxis setup has been available against Borrelia burgdorferi till now. In fact, vaccine construction using traditional methods is so expensive and time-consuming. Therefore, considering all concerns, we designed a multi-epitope-based vaccine design against Borrelia burgdorferi using in silico approaches. OBJECTIVE: To design an effective and safe vaccine that can activate cell-mediated and humoral immunity against Borrelia burgdorferi by using various bioinformatics tools. METHODS: The present study utilized different computational methodologies, covering different ideas and elements in bioinformatics tools. The protein sequence of Borrelia burgdorferi was retrieved from the NCBI database. Different B and T cell epitopes were predicated using the IEDB tool. Efficient B and T cell epitopes were further assessed for vaccine construction using linkers AAY, EAAAK and GPGPG, respectively. Furthermore, the tertiary structure of constructed vaccine was predicated, and its interaction was determined with TLR9 using ClusPro software. In addition, further atomic level detail of docked complex and their immune response were further determined by MD simulation and C-ImmSim tool, respectively. RESULTS: A protein with immunogenic potential and good vaccine properties (candidate) was identified based on high binding scores, low percentile rank, non-allergenicity and good immunological properties, which were further used to calculate epitopes. Additionally, molecular docking possesses strong interaction; seventeen H-bonds interactions were reported, such as THR101-GLU264, THR185-THR270, ARG 257-ASP210, ARG 257-ASP 210, ASP259-LYS 174, ASN263-GLU237, CYS 265-GLU 233, CYS 265-TYR 197, GLU267- THR202, GLN 270-THR202, TYR345-ASP 210, TYR345-THR 213, ARG 346-ASN209, SER350- GLU141, SER350-GLU141, ASP 424-ARG220 and ARG426-THR216 with TLR-9. Finally, high expression was determined in E. coli (CAI = (0.9045), and GC content = (72%)). Using the IMOD server, all-atom MD simulations of docked complex affirmed its significant stability. The outcomes of immune simulation indicate that both T and B cells represent a strong response to the vaccination component. CONCLUSION: This type of in-silico technique may precisely decrease valuable time and expenses in vaccine designing against Borrelia burgdorferi for experimental planning in laboratories. Currently, scientists frequently utilize bioinformatics approaches that speed up their vaccine-based lab work.


Subject(s)
Borrelia burgdorferi , Vaccines , Humans , Epitopes, T-Lymphocyte/chemistry , Molecular Docking Simulation , Vaccinology/methods , Escherichia coli , Epitopes, B-Lymphocyte/chemistry , Cloning, Molecular , Computational Biology , Vaccines, Subunit/chemistry
9.
J Mol Model ; 29(4): 99, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36928431

ABSTRACT

CONTEXT: Leishmaniasis is a group of vector-borne infectious diseases caused by over 20 pathogenic Leishmania species that are endemic in many tropical and subtropical countries. The emergence of drug-resistant strains, the adverse side effects of anti-Leishmania drugs, and the absence of a preventative vaccination strategy threaten the sensitive population. Recently, many groups of researchers have exploited the field of reverse vaccinology to develop vaccines, focusing chiefly on inducing immunity against either visceral or cutaneous leishmaniasis. METHODS: This present work involves retrieving twelve experimentally validated leishmanial antigenic protein sequences from the UniProt database, followed by their antigenicity profiling employing ANTIGENpro and Vaxijen 2.0 servers. MHC-binding epitopes for the same were predicted using both NetCTL 1.2 and SYFPEITHI servers, while epitopes for B cell were computed using ABCpred and BepiPred 2.0 servers. The screened epitopes with significantly higher scores were utilized for designing the vaccine construct with appropriate linkers and natural adjuvant. The secondary and tertiary structures of the synthetic peptide were determined by conditional random fields, shallow neural networks, and profile-profile threading alignment with iterative structure assembly simulations, respectively. The 3-D vaccine model was validated through CASP10-tested refinement and the MolProbity web server. Molecular docking and multi-scale normal mode analysis simulation were performed to analyze the best vaccine-TLR complex. Finally, computational immune simulation findings revealed promising cellular and humoral immune responses, suggesting that the engineered chimeric peptide is a potential broad-spectrum vaccine against visceral and cutaneous leishmaniasis.


Subject(s)
Leishmania , Leishmaniasis, Cutaneous , Humans , Molecular Docking Simulation , Vaccines, Combined , Epitopes , Peptides , Vaccines, Subunit/chemistry , Epitopes, T-Lymphocyte/chemistry , Computational Biology
10.
Braz J Biol ; 84: e269313, 2023.
Article in English | MEDLINE | ID: mdl-36790301

ABSTRACT

The emergence of antibiotic resistance (AR) in bacteria is becoming an alarming health concern because it allows them to adapt themselves to changing environments. It is possible to prevent the spread of AR in many ways, such as reducing antibiotic misuse in human and veterinary medicine. Streptococcus pseudopneumoniae is one of these AR bacterial species that can cause pneumonia in humans and is responsible for high mortality and morbidity rates. It is oval shaped gram-positive bacterium that shows resistance to several antibiotics like penicillin, tetracycline, ciprofloxacin, erythromycin, and co-trimoxazale and no approved vaccine is available to overcome diseases of the pathogen. Thus, substantial efforts are necessary to select protective antigens from a whole genome of pathogens that are easily tested experimentally. The in silico designed vaccine was safe and potent in immunizing individuals against the aforementioned pathogens. Herein, we utilized a subtractive genomic approach to identify potential epitope-based vaccine candidates against S. pseudopneumoniae. In total, 50850 proteins were retrieved from the NCBI, representing the complete genome of S. pseudopneumoniae. Out of the total, CD-HIT analysis identified 1022 proteins as non-redundant and 49828 proteins as redundant and further subjected for subcellular localization in which bulk of proteins was located in the cytoplasm, with seven extracellular proteins (penicillin-binding protein, alpha-amylase, solute-binding protein, hypothetical protein, CHAP domain-containing protein, polysaccharide deacetylase family protein, hypothetical protein). Six immune cells epitopes (SNLQSENDRL, RNDSLQKQAR, NPTTTSEGF, KVKKKNNKK, AYSQGSQKEH, and SVVDQVSGDF) were predicted with the help of the IEDB server. To design a multi-epitopes vaccine these immune cell epitopes were together by GPGPG and adjuvant linker to enhance immune response efficacy. The 3D structure of the designed vaccine was modeled and conducted molecular docking and dynamic simulation studies were to check the binding efficacy with immune cells receptor and dynamic behavior of the docked complex. Finally, we concluded that the designed vaccine construct can provoke a proper and protective immune response against S. pseudopneumoniae.


Subject(s)
Epitopes, T-Lymphocyte , Streptococcus , Humans , Molecular Docking Simulation , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Vaccines, Subunit/chemistry , Anti-Bacterial Agents/pharmacology
11.
Med Oncol ; 40(3): 105, 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36823384

ABSTRACT

Immunotherapy is revamping the therapeutic strategies for TNBC owing to its higher mutational burden and tumour-associated antigens. One of the most intriguing developments in cancer immunotherapy is the focus on peptide-based cancer vaccines. Thus, the current work aims to develop an efficient peptide-based vaccine against TNBC that targets Sema4A, which has recently been identified as a major regulator of TNBC progression. Initially, the antigenic peptides derived from Sema4A were determined and evaluated based on their capability to provoke immunological responses. The assessed epitopes were then linked with a suitable adjuvant (RpfB and RpfE) and appropriate linkers (AAY, GPGPG, KK and EAAAK) to preclude junctional immunogenicity. Eventually, docking and dynamics simulations are performed against TLR-2, TLR-4, TLR-7 and TLR-9 to assess the interaction between the vaccine construct and TLR receptors, as the TLR signalling pathway is critical in the host immune response. The developed vaccine was then exposed to in silico cloning and immune simulation analysis. The findings suggest that the designed vaccine could potentially evoke significant humoral and cellular immune responses in the intended organism. Considering these outcomes, the final multi-epitope vaccine could be employed to serve as an effective choice for TNBC management and may open new avenues for further studies.


Subject(s)
Semaphorins , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/therapy , Epitopes, T-Lymphocyte/chemistry , Computer Simulation , Vaccines, Subunit/chemistry , Peptides , Molecular Docking Simulation , Computational Biology , Epitopes, B-Lymphocyte/chemistry
12.
BMC Bioinformatics ; 24(1): 63, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36823524

ABSTRACT

BACKGROUND: Non-typhoidal Salmonella (NTS) is one of the important bacteria that cause foodborne diseases and invasive infections in children and elderly people. Since NTS infection is difficult to control due to the emergence of antibiotic-resistant species and its adverse effect on immune response, the development of a vaccine against NTS would be necessary. This study aimed to develop a multi-epitope vaccine against the most prevalent serovars of NTS (Salmonella Typhimurium, Salmonella Enteritidis) using an immunoinformatics approach and targeting OmpA, OmpD, and enterotoxin (Stn). RESULTS: Initially, the B cell and T cell epitopes were predicted. Then, epitopes and suitable adjuvant were assembled by molecular linkers to construct a multi-epitope vaccine. The computational tools predicted the tertiary structure, refined the tertiary structure and validated the final vaccine construct. The effectiveness of the vaccine was evaluated via molecular docking, molecular dynamics simulation, and in silico immune simulation. The vaccine model had good binding affinity and stability with MHC-I, MHC-II, and toll-like receptors (TLR-1, 2, 4) as well as activation of T cells, IgM, IgG, IFN-γ and IL-2 responses. Furthermore, after codon optimization of the vaccine sequence, this sequence was cloned in E. coli plasmid vector pET-30a (+) within restriction sites of HindIII and BamHI. CONCLUSIONS: This study, for the first time, introduced a multi-epitope vaccine based on OmpA, OmpD and enterotoxin (Stn) of NTS that could stimulate T and B cell immune responses and produced in the prokaryotic system. This vaccine was validated in-silico phase which is an essential study to reduce challenges before in vitro and in vivo studies.


Subject(s)
Bacterial Vaccines , Enterotoxins , Salmonella Infections , Humans , Bacterial Vaccines/chemistry , Bacterial Vaccines/immunology , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte/chemistry , Escherichia coli , Molecular Docking Simulation , Molecular Dynamics Simulation , Salmonella Infections/prevention & control , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
13.
BMC Bioinformatics ; 24(1): 67, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36829109

ABSTRACT

BACKGROUND: Streptococcus pneumoniae (Pneumococcus) has remained a leading cause of fatal infections such as pneumonia, meningitis, and sepsis. Moreover, this pathogen plays a major role in bacterial co-infection in patients with life-threatening respiratory virus diseases such as influenza and COVID-19. High morbidity and mortality in over one million cases, especially in very young children and the elderly, are the main motivations for pneumococcal vaccine development. Due to the limitations of the currently marketed polysaccharide-based vaccines, non-serotype-specific protein-based vaccines have received wide research interest in recent years. One step further is to identify high antigenic regions within multiple highly-conserved proteins in order to develop peptide vaccines that can affect various stages of pneumococcal infection, providing broader serotype coverage and more effective protection. In this study, immunoinformatics tools were used to design an effective multi-epitope vaccine in order to elicit neutralizing antibodies against multiple strains of pneumococcus. RESULTS: The B- and T-cell epitopes from highly protective antigens PspA (clades 1-5) and PhtD were predicted and immunodominant peptides were linked to each other with proper linkers. The domain 4 of Ply, as a potential TLR4 agonist adjuvant candidate, was attached to the end of the construct to enhance the immunogenicity of the epitope vaccine. The evaluation of the physicochemical and immunological properties showed that the final construct was stable, soluble, antigenic, and non-allergenic. Furthermore, the protein was found to be acidic and hydrophilic in nature. The protein 3D-structure was built and refined, and the Ramachandran plot, ProSA-web, ERRAT, and Verify3D validated the quality of the final model. Molecular docking analysis showed that the designed construct via Ply domain 4 had a strong interaction with TLR4. The structural stability of the docked complex was confirmed by molecular dynamics. Finally, codon optimization was performed for gene expression in E. coli, followed by in silico cloning in the pET28a(+) vector. CONCLUSION: The computational analysis of the construct showed acceptable results, however, the suggested vaccine needs to be experimentally verified in laboratory to ensure its safety and immunogenicity.


Subject(s)
COVID-19 , Streptococcus pneumoniae , Child , Humans , Child, Preschool , Aged , Molecular Docking Simulation , Escherichia coli , Toll-Like Receptor 4 , Epitopes, T-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Epitopes, B-Lymphocyte , Computational Biology/methods
14.
BMC Bioinformatics ; 24(1): 65, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36829112

ABSTRACT

BACKGROUND: It seems that several members of intestinal gut microbiota like Streptococcus bovis, Bacteroides fragilis, Helicobacter pylori, Fusobacterium nucleatum, Enterococcus faecalis, Escherichia coli, Peptostreptococcus anaerobius may be considered as the causative agents of Colorectal Cancer (CRC). The present study used bioinformatics and immunoinformatics approaches to design a potential epitope-based multi-epitope vaccine to prevent CRC with optimal population coverage. METHODS: In this study, ten amino acid sequences of CRC-related pathogens were retrieved from the NCBI database. Three ABCpred, BCPREDS and LBtope online servers were considered for B cells prediction and the IEDB server for T cells (CD4+ and CD8+) prediction. Then, validation, allergenicity, toxicity and physicochemical analysis of all sequences were performed using web servers. A total of three linkers, AAY, GPGPG, and KK were used to bind CTL, HTL and BCL epitopes, respectively. In addition, the final construct was subjected to disulfide engineering, molecular docking, immune simulation and codon adaptation to design an effective vaccine production strategy. RESULTS: A total of 19 sequences of different lengths for linear B-cell epitopes, 19 and 18 sequences were considered as epitopes of CD4+ T and CD8+ cells, respectively. The predicted epitopes were joined by appropriate linkers because they play an important role in producing an extended conformation and protein folding. The final multi-epitope construct and Toll-like receptor 4 (TLR4) were evaluated by molecular docking, which revealed stable and strong binding interactions. Immunity simulation of the vaccine showed significantly high levels of immunoglobulins, helper T cells, cytotoxic T cells and INF-γ. CONCLUSION: Finally, the results showed that the designed multi-epitope vaccine could serve as an excellent prophylactic candidate against CRC-associated pathogens, but in vitro and animal studies are needed to justify our findings for its use as a possible preventive measure.


Subject(s)
Colorectal Neoplasms , Epitopes, T-Lymphocyte , Animals , Molecular Docking Simulation , Epitopes, T-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Epitopes, B-Lymphocyte , Computational Biology/methods
15.
Acta Biomater ; 158: 535-546, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36632876

ABSTRACT

Vaccination shows great promise in cancer immunotherapy. However, the induction of robust and broad therapeutic CD8 T cell immunity against tumors is challenging due to the essential heterogenicity of tumor antigen expression. Recently, bioinspired materials have reshaped the field of cancer nanomedicine. Herein, a bioinspired nanofibrous trivalent peptide hydrogel vaccine was constructed using the spontaneous supramolecular co-assembly of three antigenic epitope-conjugated peptides, which could mimic the fibrillar structure and biological function of the extracellular matrix and naturally occurring protein assembly. The hydrogel vaccine could be accurately and flexibly adjusted to load each antigenic peptide at a defined ratio, which facilitated the antigen presentation of dendritic cells and significantly improved the initiation of CD8 T cell response and the secretion of interferon-γ (IFN-γ). C57BL/6 mice were immunized with the trivalent peptide hydrogel vaccine, where it elicited a high broad-spectrum antitumor CD8 T cell response that significantly inhibited the growth of B16 tumors in the absence of additional immunoadjuvants or delivery systems. In summary, the supramolecular assembly of triple antigenic epitope-conjugated peptides offers a simple, customizable, and versatile approach for the development of cancer vaccines with remarkable therapeutic efficacy, thereby providing a highly versatile platform for the application of personalized multivalent tumor vaccines. STATEMENT OF SIGNIFICANCE: (1) We report a feasible, versatile and bioinspired approach to manufacture a multivalent peptide-based hydrogel cancer vaccine in the absence of additional adjuvants, which closely mimics immune niches, co-delivers antigen epitopes, greatly promotes antigen presentation to DCs and their subsequent homing to dLNs and elicits a broad-spectrum antitumor CD8 T cell response, resulting in significant inhibition of B16 tumor growth. (2) This feasible and efficient co-assembly strategy provides an attractive platform for engineering a range of multivalent vaccines at defined ratios to further enhance antigen-specific T cell responses. This approach may also be used for personalized immunotherapy with neo-epitopes.


Subject(s)
Cancer Vaccines , Immunotherapy , Neoplasms , Vaccines, Subunit , Animals , Mice , Adjuvants, Immunologic , Antigens, Neoplasm , Cancer Vaccines/chemistry , Cancer Vaccines/therapeutic use , CD8-Positive T-Lymphocytes , Dendritic Cells , Epitopes , Hydrogels/chemistry , Hydrogels/therapeutic use , Immunotherapy/methods , Mice, Inbred C57BL , Neoplasms/therapy , Peptides/therapeutic use , Vaccines, Subunit/chemistry , Vaccines, Subunit/therapeutic use
16.
J Infect Public Health ; 16(2): 214-232, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36603375

ABSTRACT

BACKGROUND: Lymphocytic choriomeningitis virus (LCMV) infects many individuals worldwide and causes severe infection in the immunosuppressant recipient, spontaneous abortion, and congenital disabilities in infants. OBJECTIVES: There is no specific vaccine or therapeutics available to protect against LCMV infection; thus, there is a need to design a potential vaccine to combat the virus by developing immunity in the population. Herein, we attempted to design a potent multi-epitope vaccine for LCMV using immunoinformatics methods. METHODS: The whole proteome of the virus was screened and mapped to extract immunodominant B-cell and T-cell epitopes which were fused with appropriate linkers (EAAAK, GGGS, AAY, GPGPG, and AAY), PADRE sequence (13aa) and an adjuvant (50 S ribosomal protein L7/L12) to formulate a multi-epitope vaccine ensemble. Codon adaptation and in silico cloning of the constructed vaccine were carried out using bioinformatics tools. The secondary and tertiary structure of the vaccine construct was predicted and refined. The physicochemical profile of the designed vaccine was analyzed, and the multi-epitope vaccine's potential to bind Toll-like receptors (TLR2 and TLR4) was evaluated through molecular docking and molecular dynamics simulations. Computational immune simulation of the designed vaccine antigen was performed using the C-ImmSim server. RESULTS: The designed multi-epitope-based vaccine (613 aa) comprised 26 immunodominant (six B-cell, nine cytotoxic T lymphocytes, and 11 helper T lymphocytes) epitopes and is predicted antigenic, non-toxic, non-allergen, soluble, and topographically accessible with a suitable physicochemical profile. The designed vaccine is expected to cover a broad worldwide population (96.35 %) and stimulate a robust adaptive immune response against the virus upon administration. In silico cloning of the constructed vaccine in PET28a (+) vector ensured its optimal expression in the Escherichia coli system. Molecular docking, molecular dynamics simulation, and binding free energy estimation collectively support the stability and energetically favourable interaction of the modeled vaccine-TLR2/4 complexes. CONCLUSION: The designed multi-epitope vaccine in the present study could serve as a potential vaccine candidate to protect against LMCV infection; however, the experimental validation and safety testing of the vaccine is warranted to validate the study's outcomes.


Subject(s)
Lymphocytic choriomeningitis virus , Toll-Like Receptor 2 , Humans , Lymphocytic choriomeningitis virus/genetics , Molecular Docking Simulation , Epitopes, B-Lymphocyte/genetics , Vaccines, Subunit/chemistry , Molecular Dynamics Simulation , Computational Biology/methods
17.
Comput Biol Med ; 153: 106497, 2023 02.
Article in English | MEDLINE | ID: mdl-36599210

ABSTRACT

Recent outbreak of monkeypox disease commenced in April 2022, and on May 7, the first confirmed case was reported. The world health organization then designated monkeypox disease as a public health emergency of international outrage on July 23, after it spread to 70 non-endemic nations in less than 15 days. This catastrophic viral infection encourages the development of antiviral therapeutics due to the lack of specific treatments with negligible adverse effects. This analysis developed a highly immunogenic multiepitope subunit vaccine against the monkeypox virus using an in silico translational vaccinomics technique. Highly antigenic B cell and T cell (HTL and CTL) epitopes were predicted and conjugated with the help of unique linkers. An adjuvant (ß-defensin) and a pan-HLA DR sequence were attached at the vaccine construct's N-terminal to invoke a robust immunological response. Additionally, physiochemical, allergic, toxic, and antigenic properties were anticipated. Interactions between the vaccine candidate and the TLR3 demonstrated that the vaccine candidate triggers a robust immunological response. Finally, the stability is confirmed by the molecular dynamics study. In contrast, the modified vaccine candidate's ability to produce a protective immune response were verified by an immune dynamics simulation study conducted via C-ImmSim server. This study validates the generation of B cell, Th cell, and Tc cell populations as well as the production of IFN-γ.


Subject(s)
Monkeypox virus , Mpox (monkeypox) , Humans , Molecular Docking Simulation , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Computational Biology/methods
18.
BMC Immunol ; 24(1): 1, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604615

ABSTRACT

Continuous attempts have been made to pinpoint candidate vaccine molecules and evaluate their effectiveness in order to commercialise such vaccines for the treatment of tropical fascioliasis in livestock. The pathophysiology of fascioliasis can be related to liver damage brought on by immature flukes that migrate and feed, as well as immunological reactions to chemicals produced by the parasites and alarm signals brought on by tissue damage. Future research should, in our opinion, concentrate on the biology of invasive parasites and the resulting immune responses, particularly in the early stages of infection. The goal of the current study was to use the calcium-binding proteins from F. gigantica to create a multi-epitope subunit vaccine. The adjuvant, B-cell epitopes, CTL epitopes, and HTL epitopes that make up the vaccine construct are all connected by certain linkers. The antigenicity, allergenicity, and physiochemical properties of the vaccine construct were examined. The vaccine construct was docked with toll-like receptor 2, and simulations of the molecular dynamics of the complex's stability, interaction, and dynamics were run. After performing in silico cloning and immunosimulation, it was discovered that the construct was suitable for further investigation. New vaccination technologies and adjuvant development are advancing our food safety procedures since vaccines are seen as safe and are accepted by the user community. This research is also applicable to the F. hepatica system.


Subject(s)
Fasciola , Fascioliasis , Animals , Fascioliasis/prevention & control , Calcium , Vaccines, Subunit/chemistry , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Computational Biology/methods , Molecular Docking Simulation
19.
J Mol Recognit ; 36(4): e3007, 2023 04.
Article in English | MEDLINE | ID: mdl-36700877

ABSTRACT

Staphylococcus aureus has been widely reported to be majorly responsible for causing nosocomial infections worldwide. Due to an increase in antibiotic-resistant strains, the development of an effective vaccine against the bacteria is the most viable alternative. Therefore, in the current work, an effort has been undertaken to develop a novel peptide-based vaccine construct against S aureus that can potentially evoke the B and T cell immune responses. The fibronectin-binding proteins are an attractive target as they play a prominent role in bacterial adherence and host cell invasion and are also well conserved among rapidly mutating pathogens. Therefore, highly immunogenic linear B lymphocytes (LBL), cytotoxic T lymphocytes (CTL), and helper T lymphocytes (HTL) epitopes were identified from the antigenic fibronectin-binding proteins A and B (FnBPA and FnBPB) of S aureus using immunoinformatics approaches. The selected peptides were confirmed to be non-allergenic, non-toxic, and with a high binding affinity to the majority of human leukocyte antigens (HLA) alleles. Consequently, the multi-peptide vaccine construct was developed by fusing the screened epitopes (three LBL, five CTL, and two HTL) together with the suitable adjuvant and linkers. In addition, the tertiary conformation of the peptide construct was modeled and later docked to the Toll-like receptor 2. Subsequently, a molecular dynamics simulation of 100 ns was employed to corroborate the stability of the designed vaccine-receptor complex. Besides exhibiting high immunogenicity and conformational stability, the developed vaccine was observed to possess wide population coverage of 99.51% worldwide. Additional in vivo and in vitro validation studies would certainly corroborate the designed vaccine construct to have improved prophylactic efficacy against S aureus.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Fibronectins , Vaccinology , Epitopes, T-Lymphocyte , Epitopes, B-Lymphocyte/chemistry , Vaccines, Subunit/chemistry , Molecular Docking Simulation , Computational Biology
20.
J Infect Public Health ; 16(1): 107-116, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36508944

ABSTRACT

BACKGROUND: Monkeypox virus is an enveloped DNA virus that belongs to Poxviridae family. The virus is transmitted from rodents to primates via infected body fluids, skin lesions, and respiratory droplets. After being infected with virus, the patients experience fever, myalgia, maculopapular rash, and fluid-filled blisters. It is necessary to differentiate monkeypox virus from other poxviruses during diagnosis which can be appropriately envisioned via DNA analysis from swab samples. During small outbreaks, the virus is treated with therapies administered in other orthopoxviruses infections and does not have its own specific therapy and vaccine. Consequently, in this article, two potential peptides have been designed. METHODS: For the purpose of designing a vaccine, protein sequences were retrieved followed by the prediction of B- and T-cell epitopes. Afterward, vaccine structures were predicted which were docked with toll-like receptors. The docked complexes were analyzed with iMODS. Moreover, vaccine constructs nucleotide sequences were optimized and expressed in silico. RESULTS: COP-B7R vaccine construct (V1) has antigenicity score of 0.5400, instability index of 29.33, z-score of - 2.11-, and 42.11% GC content whereas COP-A44L vaccine construct (V2) has an antigenicity score of 0.7784, instability index of 23.33, z-score of - 0.61, and 48.63% GC content. It was also observed that COP-A44L can be expressed as a soluble protein in Escherichia coli as compared to COP-B7R which requires a different expression system. CONCLUSION: The obtained results revealed that both vaccine constructs show satisfactory outcomes after in silico investigation and have significant potential to prevent the monkeypox virus. However, COP-A44L gave better results.


Subject(s)
Epitopes, B-Lymphocyte , Monkeypox virus , Animals , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Molecular Docking Simulation , Computational Biology/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...