Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.439
Filter
1.
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
2.
Front Immunol ; 15: 1356314, 2024.
Article in English | MEDLINE | ID: mdl-38840924

ABSTRACT

Introduction: Outbreaks of coronaviruses and especially the recent COVID-19 pandemic emphasize the importance of immunological research in this area to mitigate the effect of future incidents. Bioinformatics approaches are capable of providing multisided insights from virus sequencing data, although currently available software options are not entirely suitable for a specific task of mutation surveillance within immunogenic epitopes of SARS-CoV-2. Method: Here, we describe the development of a mutation tracker, EpitopeScan, a Python3 package with command line and graphical user interface tools facilitating the investigation of the mutation dynamics in SARS-CoV-2 epitopes via analysis of multiple-sequence alignments of genomes over time. We provide an application case by examining three Spike protein-derived immunodominant CD4+ T-cell epitopes restricted by HLA-DRB1*04:01, an allele strongly associated with susceptibility to rheumatoid arthritis (RA). Mutations in these peptides are relevant for immune monitoring of CD4+ T-cell responses against SARS-CoV-2 spike protein in patients with RA. The analysis focused on 2.3 million SARS-CoV-2 genomes sampled in England. Results: We detail cases of epitope conservation over time, partial loss of conservation, and complete divergence from the wild type following the emergence of the N969K Omicron-specific mutation in November 2021. The wild type and the mutated peptide represent potential candidates to monitor variant-specific CD4+ T-cell responses. EpitopeScan is available via GitHub repository https://github.com/Aleksandr-biochem/EpitopeScan.


Subject(s)
COVID-19 , Epitopes, T-Lymphocyte , Mutation , SARS-CoV-2 , Software , Spike Glycoprotein, Coronavirus , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Humans , COVID-19/immunology , COVID-19/genetics , COVID-19/virology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , CD4-Positive T-Lymphocytes/immunology , Computational Biology/methods , Immunodominant Epitopes/immunology , Immunodominant Epitopes/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/genetics , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology
3.
HLA ; 103(6): e15541, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923358

ABSTRACT

Complications due to HCMV infection or reactivation remain a challenging clinical problem in immunocompromised patients, mainly due to insufficient or absent T-cell functionality. Knowledge of viral targets is crucial to improve monitoring of high-risk patients and optimise antiviral T-cell therapy. To expand the epitope spectrum, genetically-engineered dendritic cells (DCs) and fibroblasts were designed to secrete soluble (s)HLA-A*11:01 and infected with an HCMV mutant lacking immune evasion molecules (US2-6 + 11). More than 700 HLA-A*11:01-restricted epitopes, including more than 50 epitopes derived from a broad range of HCMV open-reading-frames (ORFs) were identified by mass spectrometry and screened for HLA-A*11:01-binding using established prediction tools. The immunogenicity of the 24 highest scoring new candidates was evaluated in vitro in healthy HLA-A*11:01+/HCMV+ donors. Thus, four subdominant epitopes and one immunodominant epitope, derived from the anti-apoptotic protein UL36 and ORFL101C (A11SAL), were identified. Their HLA-A*11:01 complex stability was verified in vitro. In depth analyses revealed highly proliferative and cytotoxic memory T-cell responses against A11SAL, with T-cell responses comparable to the immunodominant HLA-A*02:01-restricted HCMVpp65NLV epitope. A11SAL-specific T cells were also detectable in vivo in immunosuppressed transplant patients and shown to be effective in an in vitro HCMV-infection model, suggesting their crucial role in inhibiting viral replication and improvement of patient's outcome. The developed in vitro pipeline is the first to utilise genetically-engineered DCs to identify naturally presented immunodominant HCMV-derived epitopes. It therefore offers advantages over in silico predictions, is transferable to other HLA alleles, and will significantly expand the repertoire of viral targets to improve therapeutic options.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Dendritic Cells , Epitopes, T-Lymphocyte , Immunodominant Epitopes , Humans , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Immunodominant Epitopes/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A11 Antigen/immunology , HLA-A11 Antigen/genetics , Fibroblasts/immunology , Fibroblasts/virology , Antigen-Presenting Cells/immunology
4.
Front Immunol ; 15: 1413893, 2024.
Article in English | MEDLINE | ID: mdl-38915396

ABSTRACT

Introduction: Trypanosoma cruzi is a protozoan parasite that causes the tropical ailment known as Chagas disease, which has its origins in South America. Globally, it has a major impact on health and is transported by insect vector that serves as a parasite. Given the scarcity of vaccines and the limited treatment choices, we conducted a comprehensive investigation of core proteomics to explore a potential reverse vaccine candidate with high antigenicity. Methods: To identify the immunodominant epitopes, T. cruzi core proteomics was initially explored. Consequently, the vaccine sequence was engineered to possess characteristics of non-allergenicity, antigenicity, immunogenicity, and enhanced solubility. After modeling the tertiary structure of the human TLR4 receptor, the binding affinities were assessed employing molecular docking and molecular dynamics simulations (MDS). Results: Docking of the final vaccine design with TLR4 receptors revealed substantial hydrogen bond interactions. A server-based methodology for immunological simulation was developed to forecast the effectiveness against antibodies (IgM + IgG) and interferons (IFN-g). The MDS analysis revealed notable levels of structural compactness and binding stability with average RMSD of 5.03 Aring;, beta-factor 1.09e+5 Å, Rg is 44.7 Aring; and RMSF of 49.50 Aring;. This is followed by binding free energies calculation. The system stability was compromised by the complexes, as evidenced by their corresponding Gibbs free energies of -54.6 kcal/mol. Discussion: Subtractive proteomics approach was applied to determine the antigenic regions of the T cruzi. Our study utilized computational techniques to identify B- and T-cell epitopes in the T. cruzi core proteome. In current study the developed vaccine candidate exhibits immunodominant features. Our findings suggest that formulating a vaccine targeting the causative agent of Chagas disease should be the initial step in its development.


Subject(s)
Chagas Disease , Molecular Docking Simulation , Molecular Dynamics Simulation , Proteome , Protozoan Vaccines , Toll-Like Receptor 4 , Trypanosoma cruzi , Trypanosoma cruzi/immunology , Chagas Disease/immunology , Chagas Disease/prevention & control , Humans , Proteome/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Protozoan Vaccines/immunology , Animals , Immunodominant Epitopes/immunology , Proteomics/methods , Antigens, Protozoan/immunology , Antigens, Protozoan/chemistry , Antibodies, Protozoan/immunology , Protozoan Proteins/immunology , Protozoan Proteins/chemistry , Vaccine Development , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry
5.
Exp Parasitol ; 262: 108772, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723846

ABSTRACT

The family Sarcocystidae includes several intracellular coccidial parasites such as Toxoplasma gondii, Neospora caninum, Sarcocystis spp. and Hammondia spp. with heteroxenous life cycles involving different parasitic stages (oocysts/sporocysts, tachyzoites and bradyzoites in tissue cysts). The aim of this work was to evaluate monoclonal antibodies (MAb) (anti NcSAG1, anti NcSAG4 and anti TgCC2) and/or polyclonal antibodies (PAb) (anti NcSAG4 and anti TgBAG1) to label specific immunodominant antigens in different parasitic stages of N. caninum (oocyst, bradyzoite and tachyzoite), T. gondii (oocyst, cyst and tachyzoite), H. heydorni (oocyst), S. cruzi (cyst and bradyzoite) and S. falcatula (sporocyst). It was observed that the MAb directed against NcSAG1 reacted exclusively with N. caninum tachyzoites. In contrast, the MAb directed against NcSAG4 did not react with any of the parasites tested at any stage. The MAb directed against NcSAG4 reacted with both N. caninum and T. gondii tachyzoites, T. gondii tissue cysts and S. cruzi tissue cysts and bradyzoites. As expected, the MAb directed against the T. gondii tissue cyst wall antigen TgCC2 reacted with T. gondii tissue cysts, N. caninum bradyzoites, but also with T. gondii and H. heydorni oocysts and S. falcatula sporocysts. Finally, the PAb directed against the T. gondii bradyzoite proteinTgBAG1 reacted with T. gondii tissue cysts, N. caninum bradyzoites, and also with S. cruzi tissue cysts and bradyzoites. These data reveal a wide range of cross-reactions between different species of protozoa and between different developmental stages, which should be taken into account in the design and evaluation of diagnostic tests, as well as in the assessment of vaccination and challenge studies.


Subject(s)
Antibodies, Monoclonal , Antigens, Protozoan , Neospora , Sarcocystis , Toxoplasma , Sarcocystis/immunology , Neospora/immunology , Animals , Antigens, Protozoan/immunology , Toxoplasma/immunology , Antibodies, Monoclonal/immunology , Mice , Sarcocystidae/immunology , Sarcocystidae/growth & development , Immunodominant Epitopes/immunology , Enzyme-Linked Immunosorbent Assay/veterinary , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Mice, Inbred BALB C , Rabbits
6.
Virulence ; 15(1): 2360133, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38803081

ABSTRACT

Norovirus (NV) infection causes acute gastroenteritis in children and adults. Upon infection with NV, specific CD8+ T cells, which play an important role in anti-infective immunity, are activated in the host. Owing to the NV's wide genotypic variability, it is challenging to develop vaccines with cross-protective abilities against infection. To aid effective vaccine development, we examined specific CD8+ T-cell responses towards viral-structural protein (VP) epitopes, which enable binding to host susceptibility receptors. We isolated peripheral blood mononuclear cells from 196 participants to screen and identify predominant core peptides towards NV main and small envelope proteins using ex vivo and in vitro intracellular cytokine staining assays. Human leukocyte antigen (HLA) restriction characteristics were detected using next-generation sequencing. Three conservative immunodominant VP-derived CD8+ T-cell epitopes, VP294-102 (TDAARGAIN), VP2153-161 (RGPSNKSSN), and VP1141-148 (FPHIIVDV), were identified and restrictively presented by HLA-Cw * 0102, HLA-Cw * 0702, and HLA-A *1101 alleles, separately. Our findings provide useful insights into the development of future vaccines and treatments for NV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Caliciviridae Infections , Capsid Proteins , Epitopes, T-Lymphocyte , Gastroenteritis , Norovirus , Humans , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Norovirus/immunology , Norovirus/genetics , Adult , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Male , Gastroenteritis/virology , Gastroenteritis/immunology , Female , Middle Aged , Young Adult , Child , Adolescent , Leukocytes, Mononuclear/immunology , Immunodominant Epitopes/immunology , Child, Preschool , Aged
7.
Clin Immunol ; 264: 110259, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768856

ABSTRACT

The gluten-free diet for celiac disease (CeD) is restrictive and often fails to induce complete symptom and/or mucosal disease remission. Central to CeD pathogenesis is the gluten-specific CD4+ T cell that is restricted by HLA-DQ2.5 in over 85% of CeD patients, making HLA-DQ2.5 an attractive target for suppressing gluten-dependent immunity. Recently, a novel anti-HLA-DQ2.5 antibody that specifically recognizes the complexes of HLA-DQ2.5 and multiple gluten epitopes was developed (DONQ52). OBJECTIVE: To assess the ability of DONQ52 to inhibit CeD patient-derived T-cell responses to the most immunogenic gluten peptides that encompass immunodominant T cell epitopes. METHODS: We employed an in vivo gluten challenge model in patients with CeD that affords a quantitative readout of disease-relevant gluten-specific T-cell responses. HLA-DQ2.5+ CeD patients consumed food containing wheat, barley, or rye for 3 days with collection of blood before (D1) and 6 days after (D6) commencing the challenge. Peripheral blood mononuclear cells were isolated and assessed in an interferon (IFN)-γ enzyme-linked immunosorbent spot assay (ELISpot) testing responses to gluten peptides encompassing a series of immunodominant T cell epitopes. The inhibitory effect of DONQ52 (4 or 40 µg/mL) was assessed and compared to pan-HLA-DQ blockade (SPVL3 antibody). RESULTS: In HLA-DQ2.5+ CeD patients, DONQ52 reduced T cell responses to all wheat gluten peptides to an equivalent or more effective degree than pan-HLA-DQ antibody blockade. It reduced T cell responses to a cocktail of the most immunodominant wheat epitopes by a median of 87% (IQR 72-92). Notably, DONQ52 also substantially reduced T-cell responses to dominant barley hordein and rye secalin derived peptides. DONQ52 had no effect on T-cell responses to non-gluten antigens. CONCLUSION: DONQ52 can significantly block HLA-DQ2.5-restricted T cell responses to the most highly immunogenic gluten peptides in CeD. Our findings support in vitro data that DONQ52 displays selectivity and broad cross-reactivity against multiple gluten peptide:HLA-DQ2.5 complexes. This work provides proof-of-concept multi-specific antibody blockade has the potential to meaningfully inhibit pathogenic gluten-specific T-cell responses in CeD and supports ongoing therapeutic development.


Subject(s)
Antibodies, Bispecific , Celiac Disease , Glutens , HLA-DQ Antigens , Humans , Celiac Disease/immunology , Glutens/immunology , HLA-DQ Antigens/immunology , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Female , Epitopes, T-Lymphocyte/immunology , Adult , Male , CD4-Positive T-Lymphocytes/immunology , Peptides/immunology , Middle Aged , T-Lymphocytes/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Immunodominant Epitopes/immunology , Diet, Gluten-Free
8.
Vaccine ; 42(17): 3733-3743, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38705805

ABSTRACT

Hand, foot, and mouth disease (HFMD) poses a significant public health threat primarily caused by four major enteroviruses: enterovirus 71 (EV71), coxsackieviruses A16, A10, and A6. Broadly protective immune responses are essential for complete protection against these major enteroviruses. In this study, we designed a new tetravalent immunogen for HFMD, validated it in silico, in vivo evaluated the immunogenicity of the DNA-based tetravalent vaccine in mice, and identified immunogenic B-cell and T-cell epitopes. A new tetravalent immunogen, VP1me, was designed based on the chimeric protein and epitope-based vaccine principles. It contains a complete EV71 VP1 protein and six reported neutralizing B-cell epitopes derived from the four major enteroviruses causing HFMD. In silico validation using multiple immunoinformatic tools indicated good attributes of the VP1me immunogen suitable for vaccine development. The VP1me-based DNA vaccine efficiently induced both humoral and cellular immune responses in BALB/cAJcl mice. A combination of in silico prediction and immunoassays enabled the identification of immunogenic linear B-cell and CD8 T-cell epitopes within the VP1me immunogen. Immunodominant linear B-cell epitopes were identified in six regions of VP1me, with one epitope located at the N-terminus of the VP1 protein (aa 9-23) regarded as a novel epitope. Interestingly, some B-cell epitopes could also induce the CD8 T-cell response, suggesting their dual functions in immune stimulation. These results lay the groundwork for further development of VP1me as a new vaccine candidate.


Subject(s)
Antibodies, Viral , Epitopes, B-Lymphocyte , Hand, Foot and Mouth Disease , Immunodominant Epitopes , Mice, Inbred BALB C , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Epitopes, B-Lymphocyte/immunology , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/immunology , Mice , Viral Vaccines/immunology , Immunodominant Epitopes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Epitopes, T-Lymphocyte/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Enterovirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Enterovirus A, Human/immunology , Enterovirus A, Human/genetics , Immunogenicity, Vaccine , Immunity, Cellular , Immunity, Humoral
9.
Cell Rep ; 43(5): 114235, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38748880

ABSTRACT

Nanoparticle vaccines displaying mosaic receptor-binding domains (RBDs) or spike (S) from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or other sarbecoviruses are used in preparedness against potential zoonotic outbreaks. Here, we describe a self-assembling nanoparticle using lumazine synthase (LuS) as the scaffold to display RBDs from different sarbecoviruses. Mosaic nanoparticles induce sarbecovirus cross-neutralizing antibodies comparable to a nanoparticle cocktail. We find mosaic nanoparticles elicit a B cell receptor repertoire using an immunodominant germline gene pair of IGHV14-3:IGKV14-111. Most of the tested IGHV14-3:IGKV14-111 monoclonal antibodies (mAbs) are broadly cross-reactive to clade 1a, 1b, and 3 sarbecoviruses. Using mAb competition and cryo-electron microscopy, we determine that a representative IGHV14-3:IGKV14-111 mAb, M2-7, binds to a conserved epitope on the RBD, largely overlapping with the pan-sarbecovirus mAb S2H97. This suggests mosaic nanoparticles expand B cell recognition of the common epitopes shared by different clades of sarbecoviruses. These results provide immunological insights into the cross-reactive responses elicited by mosaic nanoparticles against sarbecoviruses.


Subject(s)
Nanoparticles , Nanoparticles/chemistry , Animals , Humans , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Cross Reactions/immunology , Antibody Formation/immunology , COVID-19/immunology , COVID-19/virology , Protein Domains , Mice, Inbred BALB C , Multienzyme Complexes/immunology , Female , Immunodominant Epitopes/immunology
10.
Viruses ; 16(5)2024 05 10.
Article in English | MEDLINE | ID: mdl-38793639

ABSTRACT

African Swine Fever Virus (ASFV) is a large dsDNA virus that encodes at least 150 proteins. The complexity of ASFV and lack of knowledge of effector immune functions and protective antigens have hindered the development of safe and effective ASF vaccines. In this study, we constructed four Orf virus recombinant vectors expressing individual ASFV genes B602L, -CP204L, E184L, and -I73R (ORFVΔ121-ASFV-B602L, -CP204L, -E184L, and -I73R). All recombinant viruses expressed the heterologous ASFV proteins in vitro. We then evaluated the immunogenicity of the recombinants by immunizing four-week-old piglets. In two independent animal studies, we observed high antibody titers against ASFV p30, encoded by CP204L gene. Using Pepscan ELISA, we identified a linear B-cell epitope of 12 amino acids in length (Peptide 15) located in an exposed loop region of p30 as an immunodominant ASFV epitope. Additionally, antibodies elicited against ASFV p30 presented antibody-dependent cellular cytotoxicity (ADCC) activity. These results underscore the role of p30 on antibody responses elicited against ASFV and highlight an important functional epitope that contributes to p30-specific antibody responses.


Subject(s)
African Swine Fever Virus , African Swine Fever , Antibodies, Viral , Antibody-Dependent Cell Cytotoxicity , Epitopes, B-Lymphocyte , Immunodominant Epitopes , African Swine Fever Virus/immunology , African Swine Fever Virus/genetics , Animals , Swine , Antibodies, Viral/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Immunodominant Epitopes/immunology , Immunodominant Epitopes/genetics , African Swine Fever/immunology , African Swine Fever/virology , Viral Proteins/immunology , Viral Proteins/genetics , Viral Vaccines/immunology , Viral Vaccines/genetics
11.
Front Immunol ; 15: 1395870, 2024.
Article in English | MEDLINE | ID: mdl-38799422

ABSTRACT

Emerging infectious diseases represent a significant threat to global health, with West Nile virus (WNV) being a prominent example due to its potential to cause severe neurological disorders alongside mild feverish conditions. Particularly prevalent in the continental United States, WNV has emerged as a global concern, with outbreaks indicating the urgent need for effective prophylactic measures. The current problem is that the absence of a commercial vaccine against WNV highlights a critical gap in preventive strategies against WNV. This study aims to address this gap by proposing a novel, multivalent vaccine designed using immunoinformatics approaches to elicit comprehensive humoral and cellular immune responses against WNV. The objective of the study is to provide a theoretical framework for experimental scientists to formulate of vaccine against WNV and tackle the current problem by generating an immune response inside the host. The research employs reverse vaccinology and subtractive proteomics methodologies to identify NP_041724.2 polyprotein and YP_009164950.1 truncated flavivirus polyprotein NS1 as the prime antigens. The selection process for epitopes focused on B and T-cell reactivity, antigenicity, water solubility, and non-allergenic properties, prioritizing candidates with the potential for broad immunogenicity and safety. The designed vaccine construct integrates these epitopes, connected via GPGPG linkers, and supplemented with an adjuvant with the help of another linker EAAAK, to enhance immunogenicity. Preliminary computational analyses suggest that the proposed vaccine could achieve near-universal coverage, effectively targeting approximately 99.74% of the global population, with perfect coverage in specific regions such as Sweden and Finland. Molecular docking and immune simulation studies further validate the potential efficacy of the vaccine, indicating strong binding affinity with toll-like receptor 3 (TLR-3) and promising immune response profiles, including significant antibody-mediated and cellular responses. These findings present the vaccine construct as a viable candidate for further development and testing. While the theoretical and computational results are promising, advancing from in-silico predictions to a tangible vaccine requires comprehensive laboratory validation. This next step is essential to confirm the vaccine's efficacy and safety in eliciting an immune response against WNV. Through this study, we propose a novel approach to vaccine development against WNV and contribute to the broader field of immunoinformatics, showcasing the potential to accelerate the design of effective vaccines against emerging viral threats. The journey from hypothesis to practical solution embodies the interdisciplinary collaboration essential for modern infectious disease management and prevention strategies.


Subject(s)
Computational Biology , Immunodominant Epitopes , Proteome , Vaccines, Subunit , West Nile Fever , West Nile Virus Vaccines , West Nile virus , West Nile virus/immunology , Immunodominant Epitopes/immunology , Humans , Proteome/immunology , West Nile Fever/prevention & control , West Nile Fever/immunology , West Nile Fever/virology , Computational Biology/methods , West Nile Virus Vaccines/immunology , Vaccines, Subunit/immunology , Vaccine Development , Epitopes, T-Lymphocyte/immunology , Epitopes, B-Lymphocyte/immunology , Proteomics/methods , Immunoinformatics , Protein Subunit Vaccines
12.
Vaccine ; 42(18): 3857-3873, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38616437

ABSTRACT

Dengue becomes the most common life-threatening infectious arbovirus disease globally, with prevalence in the tropical and subtropical areas. The major clinical features include dengue haemorrhagic fever (DHF) and dengue shock syndrome (DSS), a condition of hypovolemic shock. Four different serotypes of the dengue virus, known as dengue virus serotype (DENV)- 1, 2, 3 and 4 can infect humans. Only one vaccine is available in the market, named Dengvaxia by Sanofi Pasteur, but there is no desired outcome of this treatment due the antibody dependent enhancement (ADE) of the multiple dengue serotypes. As of now, there is no cure against dengue disease. Our goal in this work was to create a subunit vaccine based on several epitopes that would be effective against every serotype of the dengue virus. Here, computational methods like- immunoinformatics and bioinformatics were implemented to find out possible dominant epitopes. A total of 21 epitopes were chosen using various in-silico techniques from the expected 133 major histocompatibility complex (MHC)- I and major histocompatibility complex (MHC)- II epitopes, along with 95 B-cell epitopes which were greatly conserved. Immune stimulant, non-allergenic and non-toxic immunodominant epitopes (super epitopes) with a suitable adjuvant (Heparin-Binding Hemagglutinin Adhesin, HBHA) were used to construct the vaccine. Following the physicochemical analysis, vaccine construct was docked with Toll-like receptors (TLRs) to predict the immune stimulation. Consequently, the optimal docked complex that demonstrated the least amount of ligand-receptor complex deformability was used to conduct the molecular dynamics analysis. By following the codon optimization, the final vaccine molecule was administered into an expressing vector to perform in-silico cloning. The robust immune responses were generated in the in-silico immune simulation analysis. Hence, this study provides a hope to control the dengue infections. For validation of the immune outcomes, in-vitro as well as in-vivo investigations are essential.


Subject(s)
Dengue Vaccines , Dengue Virus , Dengue , Epitopes, B-Lymphocyte , Serogroup , Dengue Vaccines/immunology , Humans , Dengue Virus/immunology , Dengue/prevention & control , Dengue/immunology , Epitopes, B-Lymphocyte/immunology , Computer Simulation , Vaccines, Subunit/immunology , Computational Biology/methods , Immunodominant Epitopes/immunology , Antibody-Dependent Enhancement/immunology , Epitopes/immunology , Antibodies, Viral/immunology
13.
Vaccine ; 42(14): 3365-3373, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38627145

ABSTRACT

The head domain of the hemagglutinin of influenza viruses plays a dominant role in the antibody response due to the presence of immunodominant antigenic sites that are the main targets of host neutralizing antibodies. For the H1 hemagglutinin, five major antigenic sites defined as Sa, Sb, Ca1, Ca2, and Cb have been described. Although previous studies have focused on defining the hierarchy of the antigenic sites of the hemagglutinin in different human cohorts, it is still unclear if the immunodominance profile of the antigenic sites might change with the antibody levels of individuals or if other demographic factors (such as exposure history, sex, or age) could also influence the importance of the antigenic sites. The major antigenic sites of influenza viruses hemagglutinins are responsible for eliciting most of the hemagglutination inhibition antibodies in the host. To determine the antibody prevalence towards each major antigenic site, we evaluated the hemagglutination inhibition against a panel of mutant H1 viruses, each one lacking one of the "classic" antigenic sites. Our results showed that the individuals from the Stop Flu NYU cohort had an immunodominant response towards the sites Sb and Ca2 of H1 hemagglutinin. A simple logistic regression analysis of the immunodominance profiles and the hemagglutination inhibition titers displayed by each donor revealed that individuals with high hemagglutination inhibition titers against the wild-type influenza virus exhibited higher probabilities of displaying an immunodominance profile dominated by Sb, followed by Ca2 (Sb > Ca2 profile), while individuals with low hemagglutination inhibition titers presented a higher chance of displaying an immunodominance profile in which Sb and Ca2 presented the same level of immunodominance (Sb = Ca2 profile). Finally, while age exhibited an influence on the immunodominance of the antigenic sites, biological sex was not related to displaying a specific immunodominance profile.


Subject(s)
Antibodies, Viral , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus , Immunodominant Epitopes , Influenza, Human , Humans , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Male , Adult , Immunodominant Epitopes/immunology , Middle Aged , Influenza, Human/immunology , Influenza, Human/prevention & control , Young Adult , Age Factors , Sex Factors , Adolescent , Cohort Studies , Aged , Antigens, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood
14.
Front Immunol ; 15: 1329013, 2024.
Article in English | MEDLINE | ID: mdl-38665908

ABSTRACT

Introduction: Subgroups of autoantibodies directed against voltage-gated potassium channel (Kv) complex components have been associated with immunotherapy-responsive clinical syndromes. The high prevalence and the role of autoantibodies directly binding Kv remain, however, controversial. Our objective was to determine Kv autoantibody binding requirements and to clarify their contribution to the observed immune response. Methods: Binding epitopes were studied in sera (n = 36) and cerebrospinal fluid (CSF) (n = 12) from a patient cohort positive for Kv1.2 but negative for 32 common neurological autoantigens and controls (sera n = 18 and CSF n = 5) by phospho and deep mutational scans. Autoantibody specificity and contribution to the observed immune response were resolved on recombinant cells, cerebellum slices, and nerve fibers. Results: 83% of the patients (30/36) within the studied cohort shared one out of the two major binding epitopes with Kv1.2-3 reactivity. Eleven percent (4/36) of the serum samples showed no binding. Fingerprinting resolved close to identical sequence requirements for both shared epitopes. Kv autoantibody response is directed against juxtaparanodal regions in peripheral nerves and the axon initial segment in central nervous system neurons and exclusively mediated by the shared epitopes. Discussion: Systematic mapping revealed two shared autoimmune responses, with one dominant Kv1.2-3 autoantibody epitope being unexpectedly prevalent. The conservation of the molecular binding requirements among these patients indicates a uniform autoantibody repertoire with monospecific reactivity. The enhanced sensitivity of the epitope-based (10/12) compared with that of the cell-based detection (7/12) highlights its use for detection. The determined immunodominant epitope is also the primary immune response visible in tissue, suggesting a diagnostic significance and a specific value for routine screening.


Subject(s)
Autoantibodies , Autoimmunity , Immunodominant Epitopes , Kv1.2 Potassium Channel , Humans , Autoantibodies/immunology , Autoantibodies/blood , Kv1.2 Potassium Channel/immunology , Immunodominant Epitopes/immunology , Female , Male , Middle Aged , Adult , Autoantigens/immunology , Epitope Mapping , Animals
15.
Nature ; 626(7997): 160-168, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38233524

ABSTRACT

Guillain-Barré syndrome (GBS) is a rare heterogenous disorder of the peripheral nervous system, which is usually triggered by a preceding infection, and causes a potentially life-threatening progressive muscle weakness1. Although GBS is considered an autoimmune disease, the mechanisms that underlie its distinct clinical subtypes remain largely unknown. Here, by combining in vitro T cell screening, single-cell RNA sequencing and T cell receptor (TCR) sequencing, we identify autoreactive memory CD4+ cells, that show a cytotoxic T helper 1 (TH1)-like phenotype, and rare CD8+ T cells that target myelin antigens of the peripheral nerves in patients with the demyelinating disease variant. We characterized more than 1,000 autoreactive single T cell clones, which revealed a polyclonal TCR repertoire, short CDR3ß lengths, preferential HLA-DR restrictions and recognition of immunodominant epitopes. We found that autoreactive TCRß clonotypes were expanded in the blood of the same patient at distinct disease stages and, notably, that they were shared in the blood and the cerebrospinal fluid across different patients with GBS, but not in control individuals. Finally, we identified myelin-reactive T cells in the nerve biopsy from one patient, which indicates that these cells contribute directly to disease pathophysiology. Collectively, our data provide clear evidence of autoreactive T cell immunity in a subset of patients with GBS, and open new perspectives in the field of inflammatory peripheral neuropathies, with potential impact for biomedical applications.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes , Guillain-Barre Syndrome , Peripheral Nerves , Peripheral Nervous System Diseases , Th1 Cells , Humans , Biopsy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Guillain-Barre Syndrome/blood , Guillain-Barre Syndrome/cerebrospinal fluid , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/immunology , HLA-DR Antigens/immunology , Immunodominant Epitopes/immunology , Myelin Sheath/immunology , Peripheral Nerves/immunology , Peripheral Nerves/pathology , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/immunology , Peripheral Nervous System Diseases/pathology , Receptors, Antigen, T-Cell/immunology , Th1 Cells/immunology , Th1 Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Immunologic Memory
16.
Nature ; 620(7972): 128-136, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37468623

ABSTRACT

Studies have demonstrated that at least 20% of individuals infected with SARS-CoV-2 remain asymptomatic1-4. Although most global efforts have focused on severe illness in COVID-19, examining asymptomatic infection provides a unique opportunity to consider early immunological features that promote rapid viral clearance. Here, postulating that variation in the human leukocyte antigen (HLA) loci may underly processes mediating asymptomatic infection, we enrolled 29,947 individuals, for whom high-resolution HLA genotyping data were available, in a smartphone-based study designed to track COVID-19 symptoms and outcomes. Our discovery cohort (n = 1,428) comprised unvaccinated individuals who reported a positive test result for SARS-CoV-2. We tested for association of five HLA loci with disease course and identified a strong association between HLA-B*15:01 and asymptomatic infection, observed in two independent cohorts. Suggesting that this genetic association is due to pre-existing T cell immunity, we show that T cells from pre-pandemic samples from individuals carrying HLA-B*15:01 were reactive to the immunodominant SARS-CoV-2 S-derived peptide NQKLIANQF. The majority of the reactive T cells displayed a memory phenotype, were highly polyfunctional and were cross-reactive to a peptide derived from seasonal coronaviruses. The crystal structure of HLA-B*15:01-peptide complexes demonstrates that the peptides NQKLIANQF and NQKLIANAF (from OC43-CoV and HKU1-CoV) share a similar ability to be stabilized and presented by HLA-B*15:01. Finally, we show that the structural similarity of the peptides underpins T cell cross-reactivity of high-affinity public T cell receptors, providing the molecular basis for HLA-B*15:01-mediated pre-existing immunity.


Subject(s)
Alleles , Asymptomatic Infections , COVID-19 , HLA-B Antigens , Humans , COVID-19/genetics , COVID-19/immunology , COVID-19/physiopathology , COVID-19/virology , Epitopes, T-Lymphocyte/immunology , Peptides/immunology , SARS-CoV-2/immunology , HLA-B Antigens/immunology , Cohort Studies , T-Lymphocytes/immunology , Immunodominant Epitopes/immunology , Cross Reactions/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology
17.
Proc Natl Acad Sci U S A ; 119(29): e2202209119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858348

ABSTRACT

Membranous nephropathy is an autoimmune kidney disease caused by autoantibodies targeting antigens present on glomerular podocytes, instigating a cascade leading to glomerular injury. The most prevalent circulating autoantibodies in membranous nephropathy are against phospholipase A2 receptor (PLA2R), a cell surface receptor. The dominant epitope in PLA2R is located within the cysteine-rich domain, yet high-resolution structure-based mapping is lacking. In this study, we define the key nonredundant amino acids in the dominant epitope of PLA2R involved in autoantibody binding. We further describe two essential regions within the dominant epitope and spacer requirements for a synthetic peptide of the epitope for drug discovery. In addition, using cryo-electron microscopy, we have determined the high-resolution structure of PLA2R to 3.4 Å resolution, which shows that the dominant epitope and key residues within the cysteine-rich domain are accessible at the cell surface. In addition, the structure of PLA2R not only suggests a different orientation of domains but also implicates a unique immunogenic signature in PLA2R responsible for inducing autoantibody formation and recognition.


Subject(s)
Antigen Presentation , Autoantibodies , Glomerulonephritis, Membranous , Immunodominant Epitopes , Receptors, Phospholipase A2 , Autoantibodies/chemistry , Binding Sites , Cryoelectron Microscopy , Cysteine/chemistry , Glomerulonephritis, Membranous/immunology , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Protein Domains , Receptors, Phospholipase A2/chemistry , Receptors, Phospholipase A2/immunology
18.
Front Immunol ; 13: 773652, 2022.
Article in English | MEDLINE | ID: mdl-35281007

ABSTRACT

Background: Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the virus responsible for the Coronavirus Disease 2019 (COVID-19) pandemic. The emergence of variants of concern (VOCs) has become one of the most pressing issues in public health. To control VOCs, it is important to know which COVID-19 convalescent sera have cross-neutralizing activity against VOCs and how long the sera maintain this protective activity. Methods: Sera of patients infected with SARS-CoV-2 from March 2020 to January 2021 and admitted to Hyogo Prefectural Kakogawa Medical Center were selected. Blood was drawn from patients at 1-3, 3-6, and 6-8 months post onset. Then, a virus neutralization assay against SARS-CoV-2 variants (D614G mutation as conventional strain; B.1.1.7, P.1, and B.1.351 as VOCs) was performed using authentic viruses. Results: We assessed 97 sera from 42 patients. Sera from 28 patients showed neutralizing activity that was sustained for 3-8 months post onset. The neutralizing antibody titer against D614G significantly decreased in sera of 6-8 months post onset compared to those of 1-3 months post onset. However, the neutralizing antibody titers against the three VOCs were not significantly different among 1-3, 3-6, and 6-8 months post onset. Discussion: Our results indicate that neutralizing antibodies that recognize the common epitope for several variants may be maintained for a long time, while neutralizing antibodies having specific epitopes for a variant, produced in large quantities immediately after infection, may decrease quite rapidly.


Subject(s)
COVID-19/immunology , SARS-CoV-2/physiology , Aged , Antibodies, Viral/blood , Broadly Neutralizing Antibodies , Cross Reactions , Female , Humans , Immunity, Humoral , Immunodominant Epitopes/immunology , Male , Middle Aged , Time Factors
19.
Front Immunol ; 13: 822210, 2022.
Article in English | MEDLINE | ID: mdl-35173732

ABSTRACT

In HIV infection, some closely associated human leukocyte antigen (HLA) alleles are correlated with distinct clinical outcomes although presenting the same HIV epitopes. The mechanism that underpins this observation is still unknown, but may be due to the essential features of HLA alleles or T cell receptors (TCR). In this study, we investigate how T18A TCR, which is beneficial for a long-term control of HIV in clinic, recognizes immunodominant Gag epitope TL9 (TPQDLTML180-188) from HIV in the context of the antigen presenting molecule HLA-B*81:01. We found that T18A TCR exhibits differential recognition for TL9 restricted by HLA-B*81:01. Furthermore, via structural and biophysical approaches, we observed that TL9 complexes with HLA-B*81:01 undergoes no conformational change after TCR engagement. Remarkably, the CDR3ß in T18A complexes does not contact with TL9 at all but with intensive contacts to HLA-B*81:01. The binding kinetic data of T18A TCR revealed that this TCR can recognize TL9 epitope and several mutant versions, which might explain the correlation of T18A TCR with better clinic outcomes despite the relative high mutation rate of HIV. Collectively, we provided a portrait of how CD8+ T cells engage in HIV-mediated T cell response.


Subject(s)
HIV-1/metabolism , HLA-B Antigens/immunology , Mutation/genetics , Receptors, Antigen, T-Cell/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , CD8-Positive T-Lymphocytes/immunology , Humans , Immunodominant Epitopes/immunology , gag Gene Products, Human Immunodeficiency Virus/chemistry
20.
Cell Rep ; 38(7): 110363, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172147

ABSTRACT

Thymic atrophy reduces naive T cell production and contributes to increased susceptibility to viral infection with age. Expression of tissue-restricted antigen (TRA) genes also declines with age and has been thought to increase autoimmune disease susceptibility. We find that diminished expression of a model TRA gene in aged thymic stromal cells correlates with impaired clonal deletion of cognate T cells recognizing an autoantigen involved in atherosclerosis. Clonal deletion in the polyclonal thymocyte population is also perturbed. Distinct age-associated defects in the generation of antigen-specific T cells include a conspicuous decline in generation of T cells recognizing an immunodominant influenza epitope. Increased catalase activity delays thymic atrophy, and here, we show that it mitigates declining production of influenza-specific T cells and their frequency in lung after infection, but does not reverse declines in TRA expression or efficient negative selection. These results reveal important considerations for strategies to restore thymic function.


Subject(s)
Aging/immunology , Antigens/immunology , Immunity , Self Tolerance/immunology , T-Lymphocytes/immunology , Animals , Antioxidants/pharmacology , Apolipoproteins B/metabolism , Atrophy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Catalase/metabolism , Dietary Supplements , Immunity/drug effects , Immunodominant Epitopes/immunology , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae/drug effects , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Oxidation-Reduction , Oxidative Stress/drug effects , Self Tolerance/drug effects , Stromal Cells/drug effects , Stromal Cells/enzymology , T-Lymphocytes/drug effects , Thymus Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...