Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67.015
Filter
1.
Virology ; 598: 110187, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094503

ABSTRACT

Recombinant SARS-CoV-2 S protein expression was examined in Vero cells by imaging using the human monoclonal antibody panel (PD4, PD5, sc23, and sc29). The PD4 and sc29 antibodies recognised conformational specific epitopes in the S2 protein subunit at the Endoplasmic reticulum and Golgi complex. While PD5 and sc23 detected conformationally specific epitopes in the S1 protein subunit at the Golgi complex, only PD5 recognised the receptor binding domain (RBD). A comparison of the staining patterns of PD5 with non-conformationally specific antibodies that recognises the S1 subunit and RBD suggested the PD5 recognised a conformational structure within the S1 protein subunit. Our data suggests the antibody binding epitopes recognised by the human monoclonal antibodies formed at different locations in the secretory pathway during S protein transport, but a conformational change in the S1 protein subunit at the Golgi complex formed antibody binding epitopes that are recognised by virus neutralising antibodies.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Golgi Apparatus , Protein Conformation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Golgi Apparatus/metabolism , Chlorocebus aethiops , Animals , Vero Cells , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Humans , Antibodies, Neutralizing/immunology , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Epitopes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antibodies, Monoclonal/immunology , COVID-19/immunology , COVID-19/virology
2.
PLoS One ; 19(8): e0308330, 2024.
Article in English | MEDLINE | ID: mdl-39116074

ABSTRACT

The immunogenicity of cancer cells is influenced by several factors, including the expression of the major histocompatibility complex class I (MHC-I), antigen expression, and the repertoire of proteasome-produced epitope peptides. The malignant pleural mesothelioma cell line ACC-MEOS-4 (MESO-4) expresses high levels of MHC-I and Wilms tumor 1 (WT1) tumor antigens. Using a functional T cell reporter assay specific for the HLA-A*24:02 restricted WT1 epitope (WT1235, CMTWNQMNL), we searched for factors that augmented the immunogenicity of MESO-4, focusing on proteasomes, which have a central role in the antigen processing machinery. ONX-0914, a selective inhibitor of the immunoproteasome subunit ß5i, enhanced immunogenicity dose-dependently at low concentrations without cytotoxicity. In addition, CD8+ T lymphocytes recognizing WT1 showed greater cytotoxicity against MESO-4 pre-treated with ONX-0914. MESO-4 expresses a standard proteasome (SP) and immunoproteasome (IP). Notably, IP has distinct catalytic activity from SP, favoring the generation of antigenic peptides with high affinity for MHC-I in antigen-presenting cells and cancer cells. In vitro, immunoproteasome digestion assay and mass spectrometry analysis showed that IP cleaved WT1235 internally after the hydrophobic residues. Importantly, this internal cleavage of the WT1235 epitope was mitigated by ONX-0914. These results suggest that ONX-0914 prevents the internal destructive cleavage of WT1235 by IP, thereby promoting the specific presentation of the WT1 epitope by MESO-4. In conclusion, selective IP inhibitors might offer a means to modulate cancer cell immunogenicity by directing the presentation of particular tumor epitopes.


Subject(s)
Mesothelioma , Proteasome Endopeptidase Complex , Proteasome Inhibitors , WT1 Proteins , Humans , Cell Line, Tumor , WT1 Proteins/immunology , Proteasome Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/immunology , Mesothelioma/immunology , Mesothelioma/drug therapy , Epitopes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , HLA-A24 Antigen/immunology , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/drug therapy , Epitopes, T-Lymphocyte/immunology , Oligopeptides
3.
Front Immunol ; 15: 1385121, 2024.
Article in English | MEDLINE | ID: mdl-39119337

ABSTRACT

Introduction: Before they can produce their own antibodies, newborns are protected from infections by transplacental transfer of maternal IgG antibodies and after birth through breast milk IgA antibodies. Rhinovirus (RV) infections are extremely common in early childhood, and while RV infections often result in only mild upper respiratory illnesses, they can also cause severe lower respiratory illnesses such as bronchiolitis and pneumonia. Methods: We used high-density peptide arrays to profile infant and maternal antibody reactivity to capsid and full proteome sequences of three human RVs - A16, B52, and C11. Results: Numerous plasma IgG and breast milk IgA RV epitopes were identified that localized to regions of the RV capsid surface and interior, and also to several non-structural proteins. While most epitopes were bound by both IgG and IgA, there were several instances where isotype-specific and RV-specific binding were observed. We also profiled 62 unique RV-C protein loop sequences characteristic of this species' capsid VP1 protein. Discussion: Many of the RV-C loop sequences were highly bound by IgG from one-year-old infants, indicating recent or ongoing active infections, or alternatively, a level of cross-reactivity among homologous RV-C sites.


Subject(s)
Antibodies, Viral , Immunoglobulin G , Milk, Human , Rhinovirus , Humans , Milk, Human/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Immunoglobulin G/immunology , Immunoglobulin G/blood , Infant , Rhinovirus/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Picornaviridae Infections/immunology , Infant, Newborn , Epitopes/immunology , Capsid Proteins/immunology , Adult
4.
Nat Commun ; 15(1): 6867, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127753

ABSTRACT

Artificial Intelligence (AI) techniques have made great advances in assisting antibody design. However, antibody design still heavily relies on isolating antigen-specific antibodies from serum, which is a resource-intensive and time-consuming process. To address this issue, we propose a Pre-trained Antibody generative large Language Model (PALM-H3) for the de novo generation of artificial antibodies heavy chain complementarity-determining region 3 (CDRH3) with desired antigen-binding specificity, reducing the reliance on natural antibodies. We also build a high-precision model antigen-antibody binder (A2binder) that pairs antigen epitope sequences with antibody sequences to predict binding specificity and affinity. PALM-H3-generated antibodies exhibit binding ability to SARS-CoV-2 antigens, including the emerging XBB variant, as confirmed through in-silico analysis and in-vitro assays. The in-vitro assays validate that PALM-H3-generated antibodies achieve high binding affinity and potent neutralization capability against spike proteins of SARS-CoV-2 wild-type, Alpha, Delta, and the emerging XBB variant. Meanwhile, A2binder demonstrates exceptional predictive performance on binding specificity for various epitopes and variants. Furthermore, by incorporating the attention mechanism inherent in the Roformer architecture into the PALM-H3 model, we improve its interpretability, providing crucial insights into the fundamental principles of antibody design.


Subject(s)
Antibodies, Viral , COVID-19 , Complementarity Determining Regions , Epitopes , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/immunology , Humans , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Complementarity Determining Regions/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , COVID-19/immunology , COVID-19/virology , Epitopes/immunology , Antibodies, Neutralizing/immunology , Artificial Intelligence
5.
Sci Rep ; 14(1): 17910, 2024 08 02.
Article in English | MEDLINE | ID: mdl-39095538

ABSTRACT

Helicobacter pylori (H. pylori) is responsible for various chronic or acute diseases, such as stomach ulcers, dyspepsia, peptic ulcers, gastroesophageal reflux, gastritis, lymphoma, and stomach cancers. Although specific drugs are available to treat the bacterium's harmful effects, there is an urgent need to develop a preventive or therapeutic vaccine. Therefore, the current study aims to create a multi-epitope vaccine against H. pylori using lipid nanoparticles. Five epitopes from five target proteins of H. pylori, namely, Urease, CagA, HopE, SabA, and BabA, were used. Immunogenicity, MHC (Major Histocompatibility Complex) bonding, allergenicity, toxicity, physicochemical analysis, and global population coverage of the entire epitopes and final construct were carefully examined. The study involved using various bioinformatic web tools to accomplish the following tasks: modeling the three-dimensional structure of a set of epitopes and the final construct and docking them with Toll-Like Receptor 4 (TLR4). In the experimental phase, the final multi-epitope construct was synthesized using the solid phase method, and it was then enclosed in lipid nanoparticles. After synthesizing the construct, its loading, average size distribution, and nanoliposome shape were checked using Nanodrop at 280 nm, dynamic light scattering (DLS), and atomic force microscope (AFM). The designed vaccine has been confirmed to be non-toxic and anti-allergic. It can bind with different MHC alleles at a rate of 99.05%. The construct loading was determined to be about 91%, with an average size of 54 nm. Spherical shapes were also observed in the AFM images. Further laboratory tests are necessary to confirm the safety and immunogenicity of the multi-epitope vaccine.


Subject(s)
Bacterial Vaccines , Computational Biology , Helicobacter pylori , Nanoparticles , Helicobacter pylori/immunology , Nanoparticles/chemistry , Bacterial Vaccines/immunology , Bacterial Vaccines/chemistry , Computational Biology/methods , Humans , Bacterial Proteins/immunology , Bacterial Proteins/chemistry , Epitopes/immunology , Epitopes/chemistry , Molecular Docking Simulation , Antigens, Bacterial/immunology , Antigens, Bacterial/chemistry , Helicobacter Infections/prevention & control , Helicobacter Infections/immunology , Toll-Like Receptor 4/immunology , Urease/immunology , Urease/chemistry , Immunoinformatics , Liposomes
6.
BMC Genomics ; 25(1): 791, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160492

ABSTRACT

Acinetobacter baumannii is a gram-negative bacillus prevalent in nature, capable of thriving under various environmental conditions. As an opportunistic pathogen, it frequently causes nosocomial infections such as urinary tract infections, bacteremia, and pneumonia, contributing to increased morbidity and mortality in clinical settings. Consequently, developing novel vaccines against Acinetobacter baumannii is of utmost importance. In our study, we identified 10 highly conserved antigenic proteins from the NCBI and UniProt databases for epitope mapping. We subsequently screened and selected 8 CTL, HTL, and LBL epitopes, integrating them into three distinct vaccines constructed with adjuvants. Following comprehensive evaluations of immunological and physicochemical parameters, we conducted molecular docking and molecular dynamics simulations to assess the efficacy and stability of these vaccines. Our findings indicate that all three multi-epitope mRNA vaccines designed against Acinetobacter baumannii are promising; however, further animal studies are required to confirm their reliability and effectiveness.


Subject(s)
Acinetobacter baumannii , Bacterial Vaccines , Computational Biology , Acinetobacter baumannii/immunology , Acinetobacter baumannii/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Molecular Docking Simulation , Acinetobacter Infections/prevention & control , Acinetobacter Infections/immunology , Epitope Mapping , mRNA Vaccines , Molecular Dynamics Simulation , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
7.
RMD Open ; 10(3)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164049

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is characterised by the presence of autoantibodies, among which those targeting the constant region of immunoglobulin G (IgG), called rheumatoid factors (RF). Despite this link, RFs can also be found in other disorders and the healthy population, which hampers its use as a diagnostic tool. We recently showed that a subset of RA-derived RFs target a distinct epitope on the IgG-Fc, a feature that is currently not used in the clinic. METHODS: We determined immunoglobulin M (IgM)-RF levels specific against an RA-associated epitope (using our engineered next-generation RF antigen 'T3-17') in a prospective cohort of 475 patients with seropositive (for IgM-RF or aCCP) arthralgia that were followed for 5 years or until the development of arthritis. RESULTS: The presence of RFs targeting T3-17 was more strongly associated with progression to arthritis in comparison to traditional RF measurements. Within the group of patients positive for T3-17 RF the risk of arthritis development was increased as compared with wild-type RF, HR=3.2 (95% CI 2.4 to 4.3) vs HR=2.2 (95% CI 1.7 to 3.0). Predictive power of T3-17 RF was improved in combination with aCCP titres, HR=6.4 (4.7-8.7) vs HR=5.1 (3.9-6.8). This combination performed better than aCCP detection on its own. CONCLUSION: The detection of disease-specific RF is feasible and seems to improve the diagnostic power of RF and should be considered to be implemented in the clinic.


Subject(s)
Arthritis, Rheumatoid , Biomarkers , Rheumatoid Factor , Humans , Rheumatoid Factor/blood , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/blood , Male , Female , Middle Aged , Aged , Autoantibodies/blood , Immunoglobulin M/blood , Prospective Studies , Adult , Disease Progression , Epitopes/immunology , Prognosis , Immunoglobulin G/blood , Immunoglobulin G/immunology
8.
Nat Commun ; 15(1): 7124, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164226

ABSTRACT

Point-of-care serological and direct antigen testing offers actionable insights for diagnosing challenging illnesses, empowering distributed health systems. Here, we report a POC-compatible serologic test for Lyme disease (LD), leveraging synthetic peptides specific to LD antibodies and a paper-based platform for rapid, and cost-effective diagnosis. Antigenic epitopes conserved across Borrelia burgdorferi genospecies, targeted by IgG and IgM antibodies, are selected to develop a multiplexed panel for detection of LD antibodies from patient sera. Multiple peptide epitopes, when combined synergistically with a machine learning-based diagnostic model achieve high sensitivity without sacrificing specificity. Blinded validation with 15 LD-positive and 15 negative samples shows 95.5% sensitivity and 100% specificity. Blind testing with the CDC's LD repository samples confirms the test accuracy, matching lab-based two-tier results, correctly differentiating between LD and look-alike diseases. This LD diagnostic test could potentially replace the cumbersome two-tier testing, improving diagnosis and enabling earlier treatment while facilitating immune monitoring and surveillance.


Subject(s)
Antibodies, Bacterial , Borrelia burgdorferi , Immunoglobulin G , Immunoglobulin M , Lyme Disease , Sensitivity and Specificity , Serologic Tests , Lyme Disease/diagnosis , Lyme Disease/immunology , Lyme Disease/blood , Lyme Disease/microbiology , Humans , Serologic Tests/methods , Borrelia burgdorferi/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Antigens, Bacterial/immunology , Machine Learning , Epitopes/immunology , Point-of-Care Testing , Point-of-Care Systems
9.
AAPS J ; 26(5): 93, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138686

ABSTRACT

The COVID-19 pandemic continues to cause infections and deaths, which are attributable to the SARS-CoV-2 Omicron variant of concern (VOC). Moderna's response to the declining protective efficacies of current SARS-CoV-2 vaccines against Omicron was to develop a bivalent booster vaccine based on the Spike (S) protein from the Wuhan and Omicron BA.4/BA.5 strains. This approach, while commendable, is unfeasible in light of rapidly emerging mutated viral strains. PubMed and Google Scholar were systematically reviewed for peer-reviewed papers up to January 2024. Articles included focused on specific themes such as the clinical history of recombinant protein vaccine development against different diseases, including COVID-19, the production of recombinant protein vaccines using different host expression systems, aspects to consider in recombinant protein vaccine development, and overcoming problems associated with large-scale recombinant protein vaccine production. In silico approaches to identify conserved and immunogenic epitopes could provide broad protection against SARS-CoV-2 VOCs but require validation in animal models. The recombinant protein vaccine development platform has shown a successful history in clinical development. Recombinant protein vaccines incorporating conserved epitopes may utilize a number of expression systems, such as yeast (Saccharomyces cerevisiae), baculovirus-insect cells (Sf9 cells), and Escherichia coli (E. coli). Current multi-epitope subunit vaccines against SARS-CoV-2 utilizing synthetic peptides are unfeasible for large-scale immunizations. Recombinant protein vaccines based on conserved and immunogenic proteins produced using E. coli offer high production yields, convenient purification, and cost-effective production of large-scale vaccine quantities capable of protecting against the SARS-CoV-2 D614G strain and its VOCs.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccines, Synthetic , Humans , COVID-19 Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Vaccines, Synthetic/immunology , Animals , Recombinant Proteins/immunology , Vaccine Development , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Epitopes/immunology , Protein Subunit Vaccines
10.
Hum Vaccin Immunother ; 20(1): 2388344, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39165108

ABSTRACT

Monoclonal neutralizing antibodies (mAbs) are considered an important prophylactic against SARS-CoV-2 infection in at-risk populations and a strategy to counteract future sarbecovirus-induced disease. However, most mAbs isolated so far neutralize only a few sarbecovirus strains. Therefore, there is a growing interest in bispecific antibodies (bsAbs) which can simultaneously target different spike epitopes and thereby increase neutralizing breadth and prevent viral escape. Here, we generate and characterize a panel of 30 novel broadly reactive bsAbs using an efficient controlled Fab-arm exchange protocol. We specifically combine some of the broadest mAbs described so far, which target conserved epitopes on the receptor binding domain (RBD). Several bsAbs show superior cross-binding and neutralization compared to the parental mAbs and cocktails against sarbecoviruses from diverse clades, including recent SARS-CoV-2 variants. BsAbs which include mAb COVA2-02 are among the most potent and broad combinations. As a result, we study the unknown epitope of COVA2-02 and show that this mAb targets a distinct conserved region at the base of the RBD, which could be of interest when designing next-generation bsAb constructs to contribute to a better pandemic preparedness.


Subject(s)
Antibodies, Bispecific , Antibodies, Neutralizing , Antibodies, Viral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Antibodies, Bispecific/immunology , Humans , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , COVID-19/immunology , COVID-19/prevention & control , Epitopes/immunology , Neutralization Tests , Animals , Antibodies, Monoclonal/immunology
11.
Emerg Microbes Infect ; 13(1): 2389095, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101691

ABSTRACT

Influenza virus infection poses a continual menace to public health. Here, we developed soluble trimeric HA ectodomain vaccines by establishing interprotomer disulfide bonds in the stem region, which effectively preserve the native antigenicity of stem epitopes. The stable trimeric H1 ectodomain proteins exhibited higher thermal stabilities in comparison with unmodified HAs and showed strong binding activities towards a panel of anti-stem cross-reactive antibodies that recognize either interprotomer or intraprotomer epitopes. Negative stain transmission electron microscopy (TEM) analysis revealed the stable trimer architecture of the interprotomer disulfide-stapled WA11#5, NC99#2, and FLD#1 proteins as well as the irregular aggregation of unmodified HA molecules. Immunizations of mice with those trimeric HA ectodomain vaccines formulated with incomplete Freund's adjuvant elicited significantly more potent cross-neutralizing antibody responses and offered broader immuno-protection against lethal infections with heterologous influenza strains compared to unmodified HA proteins. Additionally, the findings of our study indicate that elevated levels of HA stem-specific antibody responses correlate with strengthened cross-protections. Our design strategy has proven effective in trimerizing HA ectodomains derived from both influenza A and B viruses, thereby providing a valuable reference for designing future influenza HA immunogens.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Disulfides , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Antibodies, Viral/immunology , Mice , Disulfides/chemistry , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Antibodies, Neutralizing/immunology , Female , Cross Protection/immunology , Cross Reactions , Humans , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Epitopes/immunology , Epitopes/genetics , Epitopes/chemistry , Protein Multimerization , Influenza B virus/immunology , Influenza B virus/genetics , Influenza B virus/chemistry
12.
J Gen Virol ; 105(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39158563

ABSTRACT

Bovine betacoronavirus (BoCoV) is a pneumoenteric pathogen of cattle that is closely related to human coronavirus OC43. Vaccines are administered to protect against diseases caused by BoCoV, but knowledge gaps exist with regard to correlates of protection and the effect of immune evasion on driving evolution. In this study, immune epitopes were mapped onto BoCoV structural proteins, including spike and haemagglutinin esterase (HE), and then supported with targeted gene sequencing of Irish clinical isolates and selective pressure analysis. Increased prevalence of diversifying selection and amino acid changes in some mapped immune epitopes suggests that immune escape is selecting for non-synonymous mutations arising in these regions. Selection analysis and sequencing provided increased support for neutralising antibody (nAb) epitopes compared to others, suggesting that nAbs are an important arm of the immune response to BoCoV. Phylogenetic analysis of spike and HE sequences showed that Irish isolates from this study were in the European clade, except for one HE sequence that sat in the Asian/American clade, while the spike gene of this sample was in the European clade. Recombination between a European and an Asian/American isolate would give rise to such a sequence. This study has gathered evidence suggesting that pressure to evade the nAb response is contributing to BoCoV evolution.


Subject(s)
Cattle Diseases , Coronavirus Infections , Coronavirus, Bovine , Phylogeny , Selection, Genetic , Spike Glycoprotein, Coronavirus , Animals , Cattle , Coronavirus, Bovine/genetics , Coronavirus, Bovine/immunology , Coronavirus, Bovine/isolation & purification , Cattle Diseases/virology , Cattle Diseases/immunology , Ireland , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Neutralizing/immunology , Epitopes/genetics , Epitopes/immunology , Antibodies, Viral/immunology , Immune Evasion , Hemagglutinins, Viral , Viral Fusion Proteins
13.
Nature ; 632(8025): 622-629, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112696

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a severe, post-infectious sequela of SARS-CoV-2 infection1,2, yet the pathophysiological mechanism connecting the infection to the broad inflammatory syndrome remains unknown. Here we leveraged a large set of samples from patients with MIS-C to identify a distinct set of host proteins targeted by patient autoantibodies including a particular autoreactive epitope within SNX8, a protein involved in regulating an antiviral pathway associated with MIS-C pathogenesis. In parallel, we also probed antibody responses from patients with MIS-C to the complete SARS-CoV-2 proteome and found enriched reactivity against a distinct domain of the SARS-CoV-2 nucleocapsid protein. The immunogenic regions of the viral nucleocapsid and host SNX8 proteins bear remarkable sequence similarity. Consequently, we found that many children with anti-SNX8 autoantibodies also have cross-reactive T cells engaging both the SNX8 and the SARS-CoV-2 nucleocapsid protein epitopes. Together, these findings suggest that patients with MIS-C develop a characteristic immune response to the SARS-CoV-2 nucleocapsid protein that is associated with cross-reactivity to the self-protein SNX8, demonstrating a mechanistic link between the infection and the inflammatory syndrome, with implications for better understanding a range of post-infectious autoinflammatory diseases.


Subject(s)
Antibodies, Viral , Autoantibodies , COVID-19 , Cross Reactions , Epitopes , Molecular Mimicry , SARS-CoV-2 , Systemic Inflammatory Response Syndrome , Child , Humans , Antibodies, Viral/immunology , Autoantibodies/immunology , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/immunology , COVID-19/immunology , COVID-19/virology , COVID-19/complications , Cross Reactions/immunology , Epitopes/immunology , Epitopes/chemistry , Molecular Mimicry/immunology , Phosphoproteins/chemistry , Phosphoproteins/immunology , SARS-CoV-2/chemistry , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Sorting Nexins/chemistry , Sorting Nexins/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Systemic Inflammatory Response Syndrome/virology , T-Lymphocytes/immunology
14.
Nat Commun ; 15(1): 7009, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39147753

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging bunyavirus that causes severe viral hemorrhagic fever and thrombocytopenia syndrome with a fatality rate of up to 30%. No licensed vaccines or therapeutics are currently available for humans. Here, we develop seven monoclonal antibodies (mAbs) against SFTSV surface glycoprotein Gn. Mechanistic studies show that three neutralizing mAbs (S2A5, S1G3, and S1H7) block multiple steps during SFTSV infection, including viral attachment and membrane fusion, whereas another neutralizing mAb (B1G11) primarily inhibits the viral attachment step. Epitope binning and X-ray crystallographic analyses reveal four distinct antigenic sites on Gn, three of which have not previously been reported, corresponding to domain I, domain II, and spanning domain I and domain II. One of the most potent neutralizing mAbs, S2A5, binds to a conserved epitope on Gn domain I and broadly neutralizes infection of six SFTSV strains corresponding to genotypes A to F. A single dose treatment of S2A5 affords both pre- and post-exposure protection of mice against lethal SFTSV challenge without apparent weight loss. Our results support the importance of glycoprotein Gn for eliciting a robust humoral response and pave a path for developing prophylactic and therapeutic antibodies against SFTSV infection.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Animals , Phlebovirus/immunology , Mice , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Severe Fever with Thrombocytopenia Syndrome/immunology , Severe Fever with Thrombocytopenia Syndrome/virology , Severe Fever with Thrombocytopenia Syndrome/prevention & control , Humans , Epitopes/immunology , Female , Mice, Inbred BALB C , Viral Envelope Proteins/immunology , Crystallography, X-Ray , Chlorocebus aethiops , Glycoproteins/immunology , Vero Cells
15.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39133098

ABSTRACT

Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.


Subject(s)
Computational Biology , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Sepsis/prevention & control , Sepsis/immunology , Sepsis/microbiology , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Pneumonia/prevention & control , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Vaccines/immunology , Bacterial Vaccines/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
16.
HLA ; 104(2): e15628, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39132721

ABSTRACT

Complement mediated interference with the detection of antibodies targeting HLA is a known limitation of the single antigen bead (SAB) Luminex assay. Ethylenediaminetetraacetic acid (EDTA) is currently the serum treatment of choice in most histocompatibility laboratories to block complement activation by chelating calcium. The purpose of this study was to investigate a serum with an antibody reactivity to HLA-DQ6, 7, 8 and 9 molecules, in the Luminex SAB assay, that was inhibited by treatment with EDTA. Serum was from a 55-year-old highly sensitised female renal transplant candidate that contained, among others, antibodies to an epitope containing the 74EL eplet, shared by HLA-DQ6, DQ7, DQ8 and DQ9 molecules. Serum samples were treated with EDTA, dithiothreitol (DTT), or heat prior to testing by SAB assay. EDTA-treated serum was also tested after the addition of calcium chloride (CaCl2). HLA-DQ-specific antibodies were isolated by adsorption/elution method using three informative donor cells and were tested in the absence or presence of EDTA. The antibody reactivity against HLA-DQ6, DQ7, DQ8 and DQ9 in the SAB assay was significantly inhibited by treating serum and eluates with EDTA and was restored by addition of CaCl2. The study represents the first description of a calcium-dependent epitope in HLA molecules. The relevance of this finding is that the treatment of sera with EDTA could lead to false-negative reactions in the SAB assay, which may compromise virtual crossmatching.


Subject(s)
Calcium , Edetic Acid , Epitopes , HLA-DQ Antigens , Histocompatibility Testing , Humans , Edetic Acid/pharmacology , Edetic Acid/chemistry , Epitopes/immunology , Female , Histocompatibility Testing/methods , HLA-DQ Antigens/immunology , Middle Aged , Isoantibodies/immunology , Isoantibodies/blood , Kidney Transplantation
17.
Nat Commun ; 15(1): 6892, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134522

ABSTRACT

Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.


Subject(s)
Antibodies, Viral , Cryoelectron Microscopy , Epitopes , Henipavirus Infections , Nipah Virus , Single-Domain Antibodies , Nipah Virus/immunology , Humans , Animals , Henipavirus Infections/immunology , Henipavirus Infections/prevention & control , Henipavirus Infections/virology , Epitopes/immunology , Mice , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Antibodies, Viral/immunology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Female , HEK293 Cells
18.
Commun Biol ; 7(1): 979, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134636

ABSTRACT

Previous work has shown that binding of target proteins to a sparse, unbiased sample of all possible peptide sequences is sufficient to train a machine learning model that can then predict, with statistically high accuracy, target binding to any possible peptide sequence of similar length. Here, highly sequence-specific molecular recognition is explored by measuring binding of 8 monoclonal antibodies (mAbs) with specific linear cognate epitopes to an array containing 121,715 near-random sequences about 10 residues in length. Network models trained on resulting sequence-binding values are used to predict the binding of each mAb to its cognate sequence and to an in silico generated one million random sequences. The model always ranks the binding of the cognate sequence in the top 100 sequences, and for 6 of the 8 mAbs, the cognate sequence ranks in the top ten. Practically, this approach has potential utility in selecting highly specific mAbs for therapeutics or diagnostics. More fundamentally, this demonstrates that very sparse random sampling of a large amino acid sequence spaces is sufficient to generate comprehensive models predictive of highly specific molecular recognition.


Subject(s)
Antibodies, Monoclonal , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Amino Acid Sequence , Machine Learning , Epitopes/immunology , Epitopes/chemistry , Humans , Protein Binding , Binding Sites, Antibody , Computer Simulation
19.
PLoS Pathog ; 20(8): e1012438, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39141662

ABSTRACT

The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe and fatal acute respiratory disease in humans. High fatality rates and continued infectiousness remain a pressing concern for global health preparedness. Antibodies targeted at the receptor-binding domain (RBD) are major countermeasures against human viral infection. Here, we report four potent nanobodies against MERS-CoV, which are isolated from alpaca, and especially the potency of Nb14 is highest in the pseudotyped virus assay. Structural studies show that Nb14 framework regions (FRs) are mainly involved in interactions targeting a novel epitope, which is entirely distinct from all previously reported antibodies, and disrupt the protein-carbohydrate interaction between residue W535 of RBD and hDPP4 N229-linked carbohydrate moiety (hDPP4-N229-glycan). Different from Nb14, Nb9 targets the cryptic face of RBD, which is distinctive from the hDPP4 binding site and the Nb14 epitope, and it induces the ß5-ß6 loop to inflect towards a shallow groove of the RBD and dampens the accommodation of a short helix of hDPP4. The particularly striking epitopes endow the two Nbs administrate synergistically in the pseudotyped MERS-CoV assays. These results not only character unprecedented epitopes for antibody recognition but also provide promising agents for prophylaxis and therapy of MERS-CoV infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Coronavirus Infections , Epitopes , Middle East Respiratory Syndrome Coronavirus , Single-Domain Antibodies , Middle East Respiratory Syndrome Coronavirus/immunology , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Humans , Epitopes/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Animals , Coronavirus Infections/immunology , Coronavirus Infections/virology , Camelids, New World/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Mice , Receptors, Virus/metabolism , Receptors, Virus/immunology
20.
PLoS Pathog ; 20(8): e1012383, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39093891

ABSTRACT

The SARS-CoV-2 virus responsible for the COVID-19 global pandemic has exhibited a striking capacity for viral evolution that drives continued evasion from vaccine and infection-induced immune responses. Mutations in the receptor binding domain of the S1 subunit of the spike glycoprotein have led to considerable escape from antibody responses, reducing the efficacy of vaccines and monoclonal antibody (mAb) therapies. Therefore, there is a need to interrogate more constrained regions of spike, such as the S2 subdomain. Here, we present a collection of S2 mAbs from two SARS-CoV-2 convalescent individuals that target multiple regions in S2, including regions outside of those commonly reported. One of the S2 mAbs, C20.119, which bound to a highly conserved epitope in the fusion peptide, was able to broadly neutralize across SARS-CoV-2 variants, SARS-CoV-1, and closely related zoonotic sarbecoviruses. The majority of the mAbs were non-neutralizing; however, many of them could mediate antibody-dependent cellular cytotoxicity (ADCC) at levels similar to the S1-targeting mAb S309 that was previously authorized for treatment of SARS-CoV-2 infections. Several of the mAbs with ADCC function also bound to spike trimers from other human coronaviruses (HCoVs), such as MERS-CoV and HCoV-HKU1. Our findings suggest S2 mAbs can target diverse epitopes in S2, including functional mAbs with HCoV and sarbecovirus breadth that likely target functionally constrained regions of spike. These mAbs could be developed for potential future pandemics, while also providing insight into ideal epitopes for eliciting a broad HCoV response.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Spike Glycoprotein, Coronavirus/immunology , Humans , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Epitopes/immunology , Pandemics , Betacoronavirus/immunology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Antibody-Dependent Cell Cytotoxicity/immunology
SELECTION OF CITATIONS
SEARCH DETAIL