Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.420
Filter
1.
PLoS One ; 19(7): e0292413, 2024.
Article in English | MEDLINE | ID: mdl-38959229

ABSTRACT

Salmonella infections pose a significant global public health concern due to the substantial expenses associated with monitoring, preventing, and treating the infection. In this study, we explored the core proteome of Salmonella to design a multi-epitope vaccine through Subtractive Proteomics and immunoinformatics approaches. A total of 2395 core proteins were curated from 30 different isolates of Salmonella (strain NZ CP014051 was taken as reference). Utilizing the subtractive proteomics approach on the Salmonella core proteome, Curlin major subunit A (CsgA) was selected as the vaccine candidate. csgA is a conserved gene that is related to biofilm formation. Immunodominant B and T cell epitopes from CsgA were predicted using numerous immunoinformatics tools. T lymphocyte epitopes had adequate population coverage and their corresponding MHC alleles showed significant binding scores after peptide-protein based molecular docking. Afterward, a multi-epitope vaccine was constructed with peptide linkers and Human Beta Defensin-2 (as an adjuvant). The vaccine could be highly antigenic, non-toxic, non-allergic, and have suitable physicochemical properties. Additionally, Molecular Dynamics Simulation and Immune Simulation demonstrated that the vaccine can bind with Toll Like Receptor 4 and elicit a robust immune response. Using in vitro, in vivo, and clinical trials, our findings could yield a Pan-Salmonella vaccine that might provide protection against various Salmonella species.


Subject(s)
Computational Biology , Epitopes, T-Lymphocyte , Proteomics , Salmonella , Proteomics/methods , Epitopes, T-Lymphocyte/immunology , Salmonella/immunology , Salmonella/genetics , Computational Biology/methods , Humans , Genomics/methods , Molecular Docking Simulation , Salmonella Vaccines/immunology , Animals , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Molecular Dynamics Simulation , Salmonella Infections/prevention & control , Salmonella Infections/immunology , Salmonella Infections/microbiology , Epitopes, B-Lymphocyte/immunology , Immunoinformatics
2.
Sci Rep ; 14(1): 15923, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987613

ABSTRACT

Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis (Mtb), which is one of the prominent reasons for the death of millions worldwide. The bacterium has a substantially higher mortality rate than other bacterial diseases, and the rapid rise of drug-resistant strains only makes the situation more concerning. Currently, the only licensed vaccine BCG (Bacillus Calmette-Guérin) is ineffective in preventing adult pulmonary tuberculosis prophylaxis and latent tuberculosis re-activation. Therefore, there is a pressing need to find novel and safe vaccines that provide robust immune defense and have various applications. Vaccines that combine epitopes from multiple candidate proteins have been shown to boost immunity against Mtb infection. This study applies an immunoinformatic strategy to generate an adequate multi-epitope immunization against Mtb employing five antigenic proteins. Potential B-cell, cytotoxic T lymphocyte, and helper T lymphocyte epitopes were speculated from the intended proteins and coupled with 50 s ribosomal L7/L12 adjuvant, and the vaccine was constructed. The vaccine's physicochemical profile demonstrates antigenic, soluble, and non-allergic. In the meantime, docking, molecular dynamics simulations, and essential dynamics analysis revealed that the multi-epitope vaccine structure interacted strongly with Toll-like receptors (TLR2 and TLR3). MM-PBSA analysis was performed to ascertain the system's intermolecular binding free energies accurately. The immune simulation was applied to the vaccine to forecast its immunogenic profile. Finally, in silico cloning was used to validate the vaccine's efficacy. The immunoinformatics analysis suggests the multi-epitope vaccine could induce specific immune responses, making it a potential candidate against Mtb. However, validation through the in-vivo study of the developed vaccine is essential to assess its efficacy and immunogenicity profile, which will assure active protection against Mtb.


Subject(s)
Computational Biology , Epitopes, T-Lymphocyte , Mycobacterium tuberculosis , Tuberculosis Vaccines , Vaccines, Subunit , Mycobacterium tuberculosis/immunology , Vaccines, Subunit/immunology , Tuberculosis Vaccines/immunology , Computational Biology/methods , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Dynamics Simulation , Molecular Docking Simulation , Antigens, Bacterial/immunology , Tuberculosis/prevention & control , Tuberculosis/immunology , Toll-Like Receptor 2/immunology , Immunoinformatics
3.
BMC Vet Res ; 20(1): 312, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38997779

ABSTRACT

BACKGROUND: Canine circovirus (CanineCV) is a single-stranded circular DNA virus that infects domestic and wild canids in many countries. CanineCV is associated with gastroenteritis and diarrhea, respiratory disease, and generalized vasculitis leading to a fatal event. The Capsid protein (Cap) is a structural protein of the virus which has high genetic variability and plays a role in the canine immune response. In this study, we cloned the full-length CanineCV Capsid gene (Cap). In-silico analyses were used to explore the genomic and amino acid variability and natural selection acting on the Cap gene. The immune relevance for T-cell and B-cell epitopes was predicted by the immunoinformatic approach. RESULTS: According to the Cap gene, our results showed that CanineCV was separated into five phylogenetic groups. The obtained CanineCV strain from this study was grouped with the previously discovered Thai strain (MG737385), as supported by a haplotype network. Entropy analyses revealed high nucleotide and amino acid variability of the Capsid region. Selection pressure analysis revealed four codons at positions 24, 50, 103, and 111 in the Cap protein evolved under diversifying selection. Prediction of B-cell epitopes exhibited four consensus sequences based on physiochemical properties, and eleven peptide sequences were predicted as T-cell epitopes. In addition, the positive selection sites were located within T-cell and B-cell epitopes, suggesting the role of the host immune system as a driving force in virus evolution. CONCLUSIONS: Our study provides knowledge of CanineCV genetic diversity, virus evolution, and potential epitopes for host cell immune response.


Subject(s)
Capsid Proteins , Circovirus , Phylogeny , Thailand , Circovirus/genetics , Capsid Proteins/genetics , Animals , Dogs , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Genetic Variation , Dog Diseases/virology , Amino Acid Sequence
4.
Cells ; 13(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38994930

ABSTRACT

B cell epitopes must be visible for recognition by cognate B cells and/or antibodies. Here, we studied that premise for known linear B cell epitopes that were collected from the Immune Epitope Database as being recognized by humans during microbial infections. We found that the majority of such known B cell epitopes are virus-specific linear B cell epitopes (87.96%), and most are located in antigens that remain enclosed in host cells and/or virus particles, preventing antibody recognition (18,832 out of 29,225 epitopes). Moreover, we estimated that only a minority (32.72%) of the virus-specific linear B cell epitopes that are found in exposed viral regions (e.g., the ectodomains of envelope proteins) are solvent accessible on intact antigens. Hence, we conclude that ample degradation/processing of viral particles and/or infected cells must occur prior to B cell recognition, thus shaping the B cell epitope repertoire.


Subject(s)
Epitopes, B-Lymphocyte , Epitopes, B-Lymphocyte/immunology , Humans , B-Lymphocytes/immunology , Antigens, Viral/immunology , Proteolysis , Viruses/immunology
5.
Biomed Res Int ; 2024: 4066641, 2024.
Article in English | MEDLINE | ID: mdl-38962403

ABSTRACT

The zoonotic viruses pose significant threats to public health. Nipah virus (NiV) is an emerging virus transmitted from bats to humans. The NiV causes severe encephalitis and acute respiratory distress syndrome, leading to high mortality rates, with fatality rates ranging from 40% to 75%. The first emergence of the disease was found in Malaysia in 1998-1999 and later in Bangladesh, Cambodia, Timor-Leste, Indonesia, Singapore, Papua New Guinea, Vietnam, Thailand, India, and other South and Southeast Asian nations. Currently, no specific vaccines or antiviral drugs are available. The potential advantages of epitope-based vaccines include their ability to elicit specific immune responses while minimizing potential side effects. The epitopes have been identified from the conserved region of viral proteins obtained from the UniProt database. The selection of conserved epitopes involves analyzing the genetic sequences of various viral strains. The present study identified two B cell epitopes, seven cytotoxic T lymphocyte (CTL) epitopes, and seven helper T lymphocyte (HTL) epitope interactions from the NiV proteomic inventory. The antigenic and physiological properties of retrieved protein were analyzed using online servers ToxinPred, VaxiJen v2.0, and AllerTOP. The final vaccine candidate has a total combined coverage range of 80.53%. The tertiary structure of the constructed vaccine was optimized, and its stability was confirmed with the help of molecular simulation. Molecular docking was performed to check the binding affinity and binding energy of the constructed vaccine with TLR-3 and TLR-5. Codon optimization was performed in the constructed vaccine within the Escherichia coli K12 strain, to eliminate the danger of codon bias. However, these findings must require further validation to assess their effectiveness and safety. The development of vaccines and therapeutic approaches for virus infection is an ongoing area of research, and it may take time before effective interventions are available for clinical use.


Subject(s)
Computer Simulation , Henipavirus Infections , Nipah Virus , Nipah Virus/immunology , Humans , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Viral Vaccines/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Computational Biology/methods , Epitopes, T-Lymphocyte/immunology , Vaccination , Molecular Docking Simulation , Viral Proteins/immunology , Viral Proteins/chemistry , Viral Proteins/genetics , Animals
6.
Front Immunol ; 15: 1392043, 2024.
Article in English | MEDLINE | ID: mdl-38962015

ABSTRACT

In the Americas, P. vivax is the predominant causative species of malaria, a debilitating and economically significant disease. Due to the complexity of the malaria parasite life cycle, a vaccine formulation with multiple antigens expressed in various parasite stages may represent an effective approach. Based on this, we previously designed and constructed a chimeric recombinant protein, PvRMC-1, composed by PvCyRPA, PvCelTOS, and Pvs25 epitopes. This chimeric protein was strongly recognized by naturally acquired antibodies from exposed population in the Brazilian Amazon. However, there was no investigation about the induced immune response of PvRMC-1. Therefore, in this work, we evaluated the immunogenicity of this chimeric antigen formulated in three distinct adjuvants: Stimune, AddaVax or Aluminum hydroxide (Al(OH)3) in BALB/c mice. Our results suggested that the chimeric protein PvRMC-1 were capable to generate humoral and cellular responses across all three formulations. Antibodies recognized full-length PvRMC-1 and linear B-cell epitopes from PvCyRPA, PvCelTOS, and Pvs25 individually. Moreover, mice's splenocytes were activated, producing IFN-γ in response to PvCelTOS and PvCyRPA peptide epitopes, affirming T-cell epitopes in the antigen. While aluminum hydroxide showed notable cellular response, Stimune and Addavax induced a more comprehensive immune response, encompassing both cellular and humoral components. Thus, our findings indicate that PvRMC-1 would be a promising multistage vaccine candidate that could advance to further preclinical studies.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Malaria, Vivax , Mice, Inbred BALB C , Plasmodium vivax , Protozoan Proteins , Animals , Plasmodium vivax/immunology , Plasmodium vivax/genetics , Mice , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Female , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Disease Models, Animal , Adjuvants, Immunologic , Immunogenicity, Vaccine , Antigens, Surface
7.
Sci Rep ; 14(1): 14048, 2024 06 18.
Article in English | MEDLINE | ID: mdl-38890454

ABSTRACT

Regarding several infectious diseases in fish, multiple vaccinations are not favorable. The chimeric multiepitope vaccine (CMEV) harboring several antigens for multi-disease prevention would enhance vaccine efficiency in terms of multiple disease prevention. Herein, the immunogens of tilapia's seven pathogens including E. tarda, F. columnare, F. noatunensis, S. iniae, S. agalactiae, A. hydrophila, and TiLV were used for CMEV design. After shuffling and annotating the B-cell epitopes, 5,040 CMEV primary protein structures were obtained. Secondary and tertiary protein structures were predicted by AlphaFold2 creating 25,200 CMEV. Proper amino acid alignment in the secondary structures was achieved by the Ramachandran plot. In silico determination of physiochemical and other properties including allergenicity, antigenicity, glycosylation, and conformational B-cell epitopes were determined. The selected CMEV (OSLM0467, OSLM2629, and OSLM4294) showed a predicted molecular weight (MW) of 70 kDa, with feasible sites of N- and O-glycosylation, and a number of potentially conformational B-cell epitope residues. Molecular docking, codon optimization, and in-silico cloning were tested to evaluate the possibility of protein expression. Those CMEVs will further elucidate in vitro and in vivo to evaluate the efficacy and specific immune response. This research will highlight the new era of vaccines designed based on in silico structural vaccine design.


Subject(s)
Epitopes, B-Lymphocyte , Fish Diseases , Molecular Docking Simulation , Tilapia , Animals , Tilapia/immunology , Fish Diseases/prevention & control , Fish Diseases/immunology , Fish Diseases/virology , Epitopes, B-Lymphocyte/immunology , Virus Diseases/prevention & control , Virus Diseases/immunology , Bacterial Vaccines/immunology , Viral Vaccines/immunology , Bacterial Infections/prevention & control , Bacterial Infections/immunology , Epitopes/immunology
8.
J Infect Public Health ; 17(7): 102470, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38865776

ABSTRACT

BACKGROUND: Poxviruses comprise a group of large double-stranded DNA viruses and are known to cause diseases in humans, livestock animals, and other animal species. The Mpox virus (MPXV; formerly Monkeypox), variola virus (VARV), and volepox virus (VPXV) are among the prevalent poxviruses of the Orthopoxviridae genera. The ongoing Mpox infectious disease pandemic caused by the Mpox virus has had a major impact on public health across the globe. To date, only limited repurposed antivirals and vaccines are available for the effective treatment of Mpox and other poxviruses that cause contagious diseases. METHODS: The present study was conducted with the primary goal of formulating multi-epitope vaccines against three evolutionary closed poxviruses i.e., MPXV, VARV, and VPXV using an integrated immunoinformatics and molecular modeling approach. DNA-dependent RNA polymerase (DdRp), a potential vaccine target of poxviruses, has been used to determine immunodominant B and T-cell epitopes followed by interactions analysis with Toll-like receptor 2 at the atomic level. RESULTS: Three multi-epitope vaccine constructs, namely DdRp_MPXV (V1), DdRp_VARV (V2), and DdRp_VPXV (V3) were designed. These vaccine constructs were found to be antigenic, non-allergenic, non-toxic, and soluble with desired physicochemical properties. Protein-protein docking and interaction profiling analysis depicts a strong binding pattern between the targeted immune receptor TLR2 and the structural models of the designed vaccine constructs, and manifested a number of biochemical bonds (hydrogen bonds, salt bridges, and non-bonded contacts). State-of-the-art all-atoms molecular dynamics simulations revealed highly stable interactions of vaccine constructs with TLR2 at the atomic level throughout the simulations on 300 nanoseconds. Additionally, the outcome of the immune simulation analysis suggested that designed vaccines have the potential to induce protective immunity against targeted poxviruses. CONCLUSIONS: Taken together, formulated next-generation polyvalent vaccines were found to have good efficacy against closely related poxviruses (MPXV, VARV, and VPXV) as demonstrated by our extensive immunoinformatics and molecular modeling evaluations; however, further experimental investigations are still needed.


Subject(s)
Computational Biology , Epitopes, T-Lymphocyte , Poxviridae , Viral Vaccines , Viral Vaccines/immunology , Poxviridae/immunology , Poxviridae/genetics , Computational Biology/methods , Epitopes, T-Lymphocyte/immunology , DNA-Directed RNA Polymerases/immunology , DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Models, Molecular , Animals , Humans , Poxviridae Infections/prevention & control , Poxviridae Infections/immunology , Poxviridae Infections/virology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation , Immunoinformatics
9.
Hum Vaccin Immunother ; 20(1): 2360338, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38857905

ABSTRACT

Staphylococcal Enterotoxin B (SEB), produced by Staphylococcus aureus (S. aureus), is a powerful superantigen that induces severe immune disruption and toxic shock syndrome (TSS) upon binding to MHC-II and TCR. Despite its significant impact on the pathogenesis of S. aureus, there are currently no specific therapeutic interventions available to counteract the mechanism of action exerted by this toxin. In this study, we have identified a human monoclonal antibody, named Hm0487, that specifically targets SEB by single-cell sequencing using PBMCs isolated from volunteers enrolled in a phase I clinical trial of the five-antigen S. aureus vaccine. X-ray crystallography studies revealed that Hm0487 exhibits high affinity for a linear B cell epitope in SEB (SEB138-147), which is located distantly from the site involved in the formation of the MHC-SEB-TCR ternary complex. Furthermore, in vitro studies demonstrated that Hm0487 significantly impacts the interaction of SEB with both receptors and the binding to immune cells, probably due to an allosteric effect on SEB rather than competing with receptors for binding sites. Moreover, both in vitro and in vivo studies validated that Hm0487 displayed efficient neutralizing efficacy in models of lethal shock and sepsis induced by either SEB or bacterial challenge. Our findings unveil an alternative mechanism for neutralizing the pathogenesis of SEB by Hm0487, and this antibody provides a novel strategy for mitigating both SEB-induced toxicity and S. aureus infection.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Enterotoxins , Enterotoxins/immunology , Enterotoxins/antagonists & inhibitors , Humans , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Animals , Crystallography, X-Ray , Staphylococcus aureus/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Epitopes, B-Lymphocyte/immunology , Mice , Shock, Septic/immunology , Shock, Septic/prevention & control , Female , Leukocytes, Mononuclear/immunology , Staphylococcal Vaccines/immunology , Antibodies, Bacterial/immunology , Superantigens/immunology
10.
Open Vet J ; 14(5): 1224-1242, 2024 May.
Article in English | MEDLINE | ID: mdl-38938443

ABSTRACT

Background: Porcine epidemic diarrhea (PED), caused by the porcine epidemic diarrhea virus (PEDV), is associated with high mortality and morbidity rates, especially in neonatal pigs. This has resulted in significant economic losses for the pig industry. PEDV genotype II-based vaccines were found to confer better immunity against both heterologous and homologous challenges; specifically, spike (S) proteins, which are known to play a significant role during infection, are ideal for vaccine development. Aim: This study aims to design a multi-epitope subunit vaccine targeting the S protein of the PEDV GIIa strain using an immunoinformatics approach. Methods: Various bioinformatics tools were used to predict HTL, CTL, and B-cell epitopes. The epitopes were connected using appropriate linkers and conjugated with the CTB adjuvant and M-ligand. The final multiepitope vaccine construct (fMEVc) was then docked to toll-like receptor 4 (TLR4). The stability of the fMEVc-TLR4 complex was then simulated using GROMACS. C-immsim was then used to predict the in vitro immune response of the fMEVc. Results: Six epitopes were predicted to induce antibody production, ten epitopes were predicted to induce CTL responses, and four epitopes were predicted to induce HTL responses. The assembled epitopes conjugated with the CTB adjuvant and M-ligand, fMEVc, is antigenic, non-allergenic, stable, and soluble. The construct showed a favorable binding affinity for TLR4, and the protein complex was shown to be stable through molecular dynamics simulations. A robust immune response was induced after immunization, as demonstrated through immune stimulation. Conclusion: In conclusion, the multi-epitope subunit vaccine construct for PEDV designed in this study exhibits promising antigenicity, stability, and immunogenicity, eliciting robust immune responses and suggesting its potential as a candidate for further vaccine development.


Subject(s)
Computational Biology , Coronavirus Infections , Porcine epidemic diarrhea virus , Spike Glycoprotein, Coronavirus , Swine Diseases , Vaccines, Subunit , Viral Vaccines , Animals , Porcine epidemic diarrhea virus/immunology , Vaccines, Subunit/immunology , Swine , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Viral Vaccines/immunology , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology , Genotype , Epitopes/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation , Immunoinformatics
11.
Toxins (Basel) ; 16(6)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38922165

ABSTRACT

Extensively drug-resistant Pseudomonas aeruginosa infections are emerging as a significant threat associated with adverse patient outcomes. Due to this organism's inherent properties of developing antibiotic resistance, we sought to investigate alternative strategies such as identifying "high value" antigens for immunotherapy-based purposes. Through extensive database mining, we discovered that numerous Gram-negative bacterial (GNB) genomes, many of which are known multidrug-resistant (MDR) pathogens, including P. aeruginosa, horizontally acquired the evolutionarily conserved gene encoding Zonula occludens toxin (Zot) with a substantial degree of homology. The toxin's genomic footprint among so many different GNB stresses its evolutionary importance. By employing in silico techniques such as proteomic-based phylogenetic tracing, in conjunction with comparative structural modeling, we discovered a highly conserved intermembrane associated stretch of 70 amino acids shared among all the GNB strains analyzed. The characterization of our newly identified antigen reveals it to be a "high value" vaccine candidate specific for P. aeruginosa. This newly identified antigen harbors multiple non-overlapping B- and T-cell epitopes exhibiting very high binding affinities and can adopt identical tertiary structures among the least genetically homologous P. aeruginosa strains. Taken together, using proteomic-driven reverse vaccinology techniques, we identified multiple "high value" vaccine candidates capable of eliciting a polarized immune response against all the P. aeruginosa genetic variants tested.


Subject(s)
Phylogeny , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/chemistry , Pseudomonas Vaccines/immunology , Pseudomonas Vaccines/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics
12.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38889932

ABSTRACT

Chlamydia trachomatis (Ct) is the most common sexually transmitted bacterial infection worldwide, potentially leading to severe pathologies including pelvic inflammatory disease, ectopic pregnancy, and tubal infertility if left untreated. Current strategies, including screening and antibiotics, have limited effectiveness due to high rates of asymptomatic cases and logistical challenges. A multiepitope prophylactic vaccine could afford long-term protection against infection. Immunoinformatic analyses were employed to design a multiepitope Chlamydia vaccine antigen. B- and T-cell epitopes from five highly conserved and immunogenic Ct antigens were predicted and selected for the vaccine design. The final construct, adjuvanted with cholera toxin A1 subunit (CTA1), was further screened for immunogenicity. CTA1-MECA (multiepitope Chlamydia trachomatis antigen) was identified as antigenic and nonallergenic. A tertiary structure was predicted, refined, and validated as a good quality model. Molecular docking exhibited strong interactions between the vaccine and toll-like receptor 4 (TLR4). Additionally, immune responses consistent with protection including IFN-γ, IgG + IgM antibodies, and T- and B-cell responses were predicted following vaccination in an immune simulation. Expression of the construct in an Escherichia coli expression vector proved efficient. To further validate the vaccine efficacy, we assessed its immunogenicity in mice. Immunization with CTA1-MECA elicited high levels of Chlamydia-specific antibodies in mucosal and systemic compartments.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Chlamydia Infections , Chlamydia trachomatis , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Chlamydia Infections/prevention & control , Chlamydia Infections/immunology , Animals , Chlamydia trachomatis/immunology , Epitopes, T-Lymphocyte/immunology , Mice , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Female , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Computer Simulation , Epitopes/immunology , Humans , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Cholera Toxin/immunology , Cholera Toxin/genetics , Disease Models, Animal
13.
Virus Res ; 346: 199412, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838820

ABSTRACT

African swine fever virus (ASFV) is a large double-stranded DNA virus with a complex structural architecture and encodes more than 150 proteins, where many are with unknown functions. E184L has been reported as one of the immunogenic ASFV proteins that may contribute to ASFV pathogenesis and immune evasion. However, the antigenic epitopes of E184L are not yet characterized. In this study, recombinant E184L protein was expressed in prokaryotic expression system and four monoclonal antibodies (mAbs), designated as 1A10, 2D2, 3H6, and 4C10 were generated. All four mAbs reacted specifically with ASFV infected cells. To identify the epitopes of the mAbs, a series of overlapped peptides of E184L were designed and expressed as maltose binding fusion proteins. Accordingly, the expressed fusion proteins were probed with each E184L mAb separately by using Western blot. Following a fine mapping, the minimal linear epitope recognized by mAb 1A10 was identified as 119IQRQGFL125, and mAbs 2D2, 3H6, and 4C10 recognized a region located between 153DPTEFF158. Alignment of amino acids of E184L revealed that the two linear epitopes are highly conserved among different ASFV isolates. Furthermore, the potential application of the two epitopes in ASFV diagnosis was assessed through epitope-based ELISA using 24 ASFV positive and 18 negative pig serum and the method were able to distinguish positive and negative samples, indicating the two epitopes are dominant antigenic sites. To our knowledge, this is the first study to characterize the B cell epitopes of the antigenic E184L protein of ASFV, offering valuable tools for future research, as well as laying a foundation for serological diagnosis and epitope-based marker vaccine development.


Subject(s)
African Swine Fever Virus , Antibodies, Monoclonal , Antibodies, Viral , Epitope Mapping , Epitopes, B-Lymphocyte , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Animals , Antibodies, Viral/immunology , Swine , African Swine Fever/immunology , African Swine Fever/virology , Mice , Viral Proteins/immunology , Viral Proteins/genetics , Viral Proteins/chemistry , Antigens, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/chemistry , Mice, Inbred BALB C
14.
Trop Biomed ; 41(1): 84-96, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38852138

ABSTRACT

Rhipicephalus microplus, known as the hard tick, is a vector for the parasites Babesia spp. and Anaplasma marginale, both of which can cause significant financial losses to the livestock industry. There is currently no effective vaccine for R. microplus tick infestations, despite the identification of numerous prospective tick vaccine candidates. As a result, the current research set out to develop an immunoinformatics-based strategy using existing methods for designing a multi-epitope based vaccination that is not only effective but also safe and capable of eliciting cellular and humoral immune responses. First, R. microplus proteins Bm86, Subolesin, and Bm95 were used to anticipate and link B and T-cell epitopes (HTL and CTL) to one another. Antigenicity testing, allergenicity assessment, and toxicity screening were just a few of the many immunoinformatics techniques used to identify potent epitopes. Multi-epitope vaccine design was chosen based on the antigenic score 0.935 that is promising vaccine candidate. Molecular docking was used to determine the nature of the interaction between TLR2 and the vaccine construct. Finally, molecular dynamic simulation was used to assess the stability and compactness of the resulting vaccination based on docking scores. The developed vaccine was shown to be stable, have immunogenic qualities, be soluble, and to have high expression by in silico cloning. These findings suggest that experimental investigation of the multi-epitope based vaccine designed in the current study will produce achievable vaccine candidates against R. microplus ticks, enabling more effective control of infestations.


Subject(s)
Arthropod Proteins , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Rhipicephalus , Vaccines , Rhipicephalus/immunology , Animals , Vaccines/immunology , Arthropod Proteins/immunology , Arthropod Proteins/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation , Tick Infestations/prevention & control , Tick Infestations/veterinary , Tick Infestations/immunology , Molecular Dynamics Simulation , Epitopes/immunology , Immunoinformatics , Antigens , Membrane Glycoproteins , Recombinant Proteins
15.
Methods Mol Biol ; 2813: 245-280, 2024.
Article in English | MEDLINE | ID: mdl-38888783

ABSTRACT

Identifying antigens within a pathogen is a critical task to develop effective vaccines and diagnostic methods, as well as understanding the evolution and adaptation to host immune responses. Historically, antigenicity was studied with experiments that evaluate the immune response against selected fragments of pathogens. Using this approach, the scientific community has gathered abundant information regarding which pathogenic fragments are immunogenic. The systematic collection of this data has enabled unraveling many of the fundamental rules underlying the properties defining epitopes and immunogenicity, and has resulted in the creation of a large panel of immunologically relevant predictive (in silico) tools. The development and application of such tools have proven to accelerate the identification of novel epitopes within biomedical applications reducing experimental costs. This chapter introduces some basic concepts about MHC presentation, T cell and B cell epitopes, the experimental efforts to determine those, and focuses on state-of-the-art methods for epitope prediction, highlighting their strengths and limitations, and catering instructions for their rational use.


Subject(s)
Computational Biology , Computer Simulation , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Humans , Epitopes, T-Lymphocyte/immunology , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes/immunology , Software , Animals , Epitope Mapping/methods , Antigen Presentation/immunology
16.
Front Cell Infect Microbiol ; 14: 1351303, 2024.
Article in English | MEDLINE | ID: mdl-38881736

ABSTRACT

Introduction: Fowl adenovirus (FAdV) is a significant pathogen in poultry, causing various diseases such as hepatitis-hydropericardium, inclusion body hepatitis, and gizzard erosion. Different serotypes of FAdV are associated with specific conditions, highlighting the need for targeted prevention strategies. Given the rising prevalence of FAdV-related diseases globally, effective vaccination and biosecurity measures are crucial. In this study, we explore the potential of structural proteins to design a multi-epitope vaccine targeting FAdV. Methods: We employed an in silico approach to design the multi-epitope vaccine. Essential viral structural proteins, including hexon, penton, and fiber protein, were selected as vaccine targets. T-cell and B-cell epitopes binding to MHC-I and MHC-II molecules were predicted using computational methods. Molecular docking studies were conducted to validate the interaction of the multi-epitope vaccine candidate with chicken Toll-like receptors 2 and 5. Results: Our in silico methodology successfully identified potential T-cell and B-cell epitopes within the selected viral structural proteins. Molecular docking studies revealed strong interactions between the multi-epitope vaccine candidate and chicken Toll-like receptors 2 and 5, indicating the structural integrity and immunogenic potential of the designed vaccine. Discussion: The designed multi-epitope vaccine presents a promising approach for combating FAdV infections in chickens. By targeting essential viral structural proteins, the vaccine is expected to induce a robust immunological response. The in silico methodology utilized in this study provides a rapid and cost-effective means of vaccine design, offering insights into potential vaccine candidates before experimental validation. Future studies should focus on in vitro and in vivo evaluations to further assess the efficacy and safety of the proposed vaccine.


Subject(s)
Adenoviridae Infections , Chickens , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation , Poultry Diseases , Vaccines, Subunit , Animals , Vaccines, Subunit/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Adenoviridae Infections/prevention & control , Adenoviridae Infections/veterinary , Adenoviridae Infections/immunology , Viral Vaccines/immunology , Viral Structural Proteins/immunology , Viral Structural Proteins/genetics , Aviadenovirus/immunology , Aviadenovirus/genetics , Computer Simulation , Protein Subunit Vaccines
17.
J Immunol Methods ; 530: 113697, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823576

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) caused by the PRRS virus (PRRSV) is one of the most severe swine diseases causing great economic losses for the international swine industry. Non-structural protein 4 (NSP4) is critical to the life cycle of PRRSV and contains dominant B cell epitopes. This study prepared a monoclonal antibody against Nsp4, and 2D11, which contained the sequence 138KQGGGIVTRPSGQFCN153, was confirmed as the epitope. A 2D11-based double antibody sandwich enzyme-linked immunosorbent assay (dasELISA) was next developed with a cut value of 0.1987. A total of 1354 pig serum samples were detected by dasELISA and compared to a commercial ELISA kit (N-coated iELISA), resulting in a positive coincidence rate of 98.8% and negative coincidence rate of 96.9%. A total of 119 sera were positive by dasELISA while negative by iELISA. Higher positive rates by dasELISA were found in pig farms where PRRSV antibody levels varied widely. These results indicated that the dasELISA was a useful tool to detect PRRSV antibody in clinical samples.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Enzyme-Linked Immunosorbent Assay , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Viral Nonstructural Proteins , Porcine respiratory and reproductive syndrome virus/immunology , Animals , Enzyme-Linked Immunosorbent Assay/methods , Swine , Antibodies, Monoclonal/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/diagnosis , Porcine Reproductive and Respiratory Syndrome/blood , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Nonstructural Proteins/immunology , Immunodominant Epitopes/immunology , Epitopes, B-Lymphocyte/immunology
18.
Viral Immunol ; 37(5): 259-265, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38848306

ABSTRACT

Cytomegalovirus (CMV) has long been thought to have an association with glioblastoma multiforme (GBM), although the exact role of CMV and any subsequent implications for treatment have yet to be fully understood. This study addressed whether IGH complementarity determining region-3 (CDR3)-CMV protein chemical complementarity, with IGH CDR3s representing both tumor resident and blood-sourced IGH recombinations, was associated with overall survival (OS) distinctions. IGH recombination sequencing reads were obtained from (a) the Clinical Proteomic Tumor Analysis Consortium, tumor RNAseq files; and (b) the cancer genome atlas, blood exome-derived files. The Adaptive Match web tool was used to calculate chemical complementarity scores (CSs) based on hydrophobic interactions, and those scores were used to group GBM cases and assess survival probabilities. We found a higher OS probability for cases whose hydrophobic IGH CDR3-CMV protein chemical complementarity scores (Hydro CSs) were in the upper 50th percentile for several CMV proteins, including UL99 and UL123, as well as for CSs based on known B cell epitopes representing these proteins. We also identified multiple immune signature genes, including CD79A and TNFRSF17, for which higher RNA expression was associated with higher Hydro CSs. Results were consistent with the idea that stronger immunoglobulin responses to CMV are associated with better OS probabilities for GBM.


Subject(s)
Complementarity Determining Regions , Cytomegalovirus Infections , Cytomegalovirus , Glioblastoma , Viral Proteins , Humans , Glioblastoma/mortality , Glioblastoma/genetics , Glioblastoma/virology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Cytomegalovirus Infections/mortality , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Viral Proteins/genetics , Viral Proteins/immunology , Immunoglobulin Heavy Chains/genetics , Female , Middle Aged , Male , Survival Analysis , Aged , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics
19.
Nucleic Acids Res ; 52(W1): W533-W539, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38742639

ABSTRACT

Prediction of conformational B-cell epitopes is a crucial task in vaccine design and development. In this work, we have developed SEMA 2.0, a user-friendly web platform that enables the research community to tackle the B-cell epitopes prediction problem using state-of-the-art protein language models. SEMA 2.0 offers comprehensive research tools for sequence- and structure-based conformational B-cell epitopes prediction, accurate identification of N-glycosylation sites, and a distinctive module for comparing the structures of antigen B-cell epitopes enhancing our ability to analyze and understand its immunogenic properties. SEMA 2.0 website https://sema.airi.net is free and open to all users and there is no login requirement. Source code is available at https://github.com/AIRI-Institute/SEMAi.


Subject(s)
Epitopes, B-Lymphocyte , Internet , Software , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Artificial Intelligence , Humans , Protein Conformation , Glycosylation
20.
Front Immunol ; 15: 1380732, 2024.
Article in English | MEDLINE | ID: mdl-38690283

ABSTRACT

Haemophilus parainfluenzae is a Gram-negative opportunist pathogen within the mucus of the nose and mouth without significant symptoms and has an ability to cause various infections ranging from ear, eye, and sinus to pneumonia. A concerning development is the increasing resistance of H. parainfluenzae to beta-lactam antibiotics, with the potential to cause dental infections or abscesses. The principal objective of this investigation is to utilize bioinformatics and immuno-informatic methodologies in the development of a candidate multi-epitope Vaccine. The investigation focuses on identifying potential epitopes for both B cells (B lymphocytes) and T cells (helper T lymphocytes and cytotoxic T lymphocytes) based on high non-toxic and non-allergenic characteristics. The selection process involves identifying human leukocyte antigen alleles demonstrating strong associations with recognized antigenic and overlapping epitopes. Notably, the chosen alleles aim to provide coverage for 90% of the global population. Multi-epitope constructs were designed by using suitable linker sequences. To enhance the immunological potential, an adjuvant sequence was incorporated using the EAAAK linker. The final vaccine construct, comprising 344 amino acids, was achieved after the addition of adjuvants and linkers. This multi-epitope Vaccine demonstrates notable antigenicity and possesses favorable physiochemical characteristics. The three-dimensional conformation underwent modeling and refinement, validated through in-silico methods. Additionally, a protein-protein molecular docking analysis was conducted to predict effective binding poses between the multi-epitope Vaccine and the Toll-like receptor 4 protein. The Molecular Dynamics (MD) investigation of the docked TLR4-vaccine complex demonstrated consistent stability over the simulation period, primarily attributed to electrostatic energy. The docked complex displayed minimal deformation and enhanced rigidity in the motion of residues during the dynamic simulation. Furthermore, codon translational optimization and computational cloning was performed to ensure the reliability and proper expression of the multi-Epitope Vaccine. It is crucial to emphasize that despite these computational validations, experimental research in the laboratory is imperative to demonstrate the immunogenicity and protective efficacy of the developed vaccine. This would involve practical assessments to ascertain the real-world effectiveness of the multi-epitope Vaccine.


Subject(s)
Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Humans , Epitopes, T-Lymphocyte/immunology , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Molecular Docking Simulation , Haemophilus Infections/prevention & control , Haemophilus Infections/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Vaccine Development
SELECTION OF CITATIONS
SEARCH DETAIL
...