Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.763
Filter
1.
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
2.
Front Immunol ; 15: 1392477, 2024.
Article in English | MEDLINE | ID: mdl-38774878

ABSTRACT

Introduction: Accumulating evidence indicates the importance of T cell immunity in vaccination-induced protection against severe COVID-19 disease, especially against SARS-CoV-2 Variants-of-Concern (VOCs) that more readily escape from recognition by neutralizing antibodies. However, there is limited knowledge on the T cell responses across different age groups and the impact of CMV status after primary and booster vaccination with different vaccine combinations. Moreover, it remains unclear whether age has an effect on the ability of T cells to cross-react against VOCs. Methods: Therefore, we interrogated the Spike-specific T cell responses in healthy adults of the Dutch population across different ages, whom received different vaccine types for the primary series and/or booster vaccination, using IFNÉ£ ELISpot. Cells were stimulated with overlapping peptide pools of the ancestral Spike protein and different VOCs. Results: Robust Spike-specific T cell responses were detected in the vast majority of participants upon the primary vaccination series, regardless of the vaccine type (i.e. BNT162b2, mRNA-1273, ChAdOx1 nCoV-19, or Ad26.COV2.S). Clearly, in the 70+ age group, responses were overall lower and showed more variation compared to younger age groups. Only in CMV-seropositive older adults (>70y) there was a significant inverse relation of age with T cell responses. Although T cell responses increased in all age groups after booster vaccination, Spike-specific T cell frequencies remained lower in the 70+ age group. Regardless of age or CMV status, primary mRNA-1273 vaccination followed by BNT162b2 booster vaccination showed limited booster effect compared to the BNT162b2/BNT162b2 or BNT162b2/mRNA-1273 primary-booster regimen. A modest reduction in cross-reactivity to the Alpha, Delta and Omicron BA.1, but not the Beta or Gamma variant, was observed after primary vaccination. Discussion: Together, this study shows that age, CMV status, but also the primary-booster vaccination regimen influence the height of the vaccination-induced Spike-specific T cell response, but did not impact the VOC cross-reactivity.


Subject(s)
COVID-19 , Cross Reactions , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , T-Lymphocytes , Humans , Cross Reactions/immunology , SARS-CoV-2/immunology , Middle Aged , Adult , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Aged , Male , T-Lymphocytes/immunology , Female , Spike Glycoprotein, Coronavirus/immunology , Age Factors , Young Adult , COVID-19 Vaccines/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Immunization, Secondary , Cytomegalovirus/immunology , BNT162 Vaccine/immunology , Vaccination , 2019-nCoV Vaccine mRNA-1273/immunology , ChAdOx1 nCoV-19/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , Aged, 80 and over
3.
Influenza Other Respir Viruses ; 18(5): e13309, 2024 May.
Article in English | MEDLINE | ID: mdl-38725111

ABSTRACT

BACKGROUND: The newly emerged SARS-CoV-2 possesses shared antigenic epitopes with other human coronaviruses. We investigated if COVID-19 vaccination or SARS-CoV-2 infection may boost cross-reactive antibodies to other human coronaviruses. METHODS: Prevaccination and postvaccination sera from SARS-CoV-2 naïve healthy subjects who received three doses of the mRNA vaccine (BioNTech, BNT) or the inactivated vaccine (CoronaVac, CV) were used to monitor the level of cross-reactive antibodies raised against other human coronaviruses by enzyme-linked immunosorbent assay. In comparison, convalescent sera from COVID-19 patients with or without prior vaccination history were also tested. Pseudoparticle neutralization assay was performed to detect neutralization antibody against MERS-CoV. RESULTS: Among SARS-CoV-2 infection-naïve subjects, BNT or CV significantly increased the anti-S2 antibodies against Betacoronaviruses (OC43 and MERS-CoV) but not Alphacoronaviruses (229E). The prevaccination antibody response to the common cold human coronaviruses did not negatively impact the postvaccination antibody response to SARS-CoV-2. Cross-reactive antibodies that binds to the S2 protein of MERS-CoV were similarly detected from the convalescent sera of COVID-19 patients with or without vaccination history. However, these anti-S2 antibodies do not possess neutralizing activity in MERS-CoV pseudoparticle neutralization tests. CONCLUSIONS: Our results suggest that SARS-CoV-2 infection or vaccination may potentially modulate population immune landscape against previously exposed or novel human coronaviruses. The findings have implications for future sero-epidemiological studies on MERS-CoV.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Cross Reactions , SARS-CoV-2 , Humans , Cross Reactions/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Adult , Male , Female , Vaccination , Middle Aged , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Neutralization Tests , Middle East Respiratory Syndrome Coronavirus/immunology , Young Adult , mRNA Vaccines/immunology
4.
Front Immunol ; 15: 1381508, 2024.
Article in English | MEDLINE | ID: mdl-38690272

ABSTRACT

Seasonal influenza remains a serious global health problem, leading to high mortality rates among the elderly and individuals with comorbidities. Vaccination is generally accepted as the most effective strategy for influenza prevention. While current influenza vaccines are effective, they still have limitations, including narrow specificity for certain serological variants, which may result in a mismatch between vaccine antigens and circulating strains. Additionally, the rapid variability of the virus poses challenges in providing extended protection beyond a single season. Therefore, mRNA technology is particularly promising for influenza prevention, as it enables the rapid development of multivalent vaccines and allows for quick updates of their antigenic composition. mRNA vaccines have already proven successful in preventing COVID-19 by eliciting rapid cellular and humoral immune responses. In this study, we present the development of a trivalent mRNA vaccine candidate, evaluate its immunogenicity using the hemagglutination inhibition assay, ELISA, and assess its efficacy in animals. We demonstrate the higher immunogenicity of the mRNA vaccine candidate compared to the inactivated split influenza vaccine and its enhanced ability to generate a cross-specific humoral immune response. These findings highlight the potential mRNA technology in overcoming current limitations of influenza vaccines and hold promise for ensuring greater efficacy in preventing seasonal influenza outbreaks.


Subject(s)
Antibodies, Viral , Cross Reactions , Immunity, Humoral , Influenza Vaccines , mRNA Vaccines , Influenza Vaccines/immunology , Animals , mRNA Vaccines/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Humans , Cross Reactions/immunology , Mice , Influenza, Human/prevention & control , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Female , Seasons , Immunogenicity, Vaccine , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Mice, Inbred BALB C , Influenza A Virus, H1N1 Subtype/immunology , COVID-19/prevention & control , COVID-19/immunology , Vaccination
5.
Viruses ; 16(5)2024 05 15.
Article in English | MEDLINE | ID: mdl-38793670

ABSTRACT

The West Nile Virus (WNV), a member of the family Flaviviridae, is an emerging mosquito-borne flavivirus causing potentially severe infections in humans and animals involving the central nervous system (CNS). Due to its emerging tendency, WNV now occurs in many areas where other flaviviruses are co-occurring. Cross-reactive antibodies with flavivirus infections or vaccination (e.g., tick-borne encephalitis virus (TBEV), Usutu virus (USUV), yellow fever virus (YFV), dengue virus (DENV), Japanese encephalitis virus (JEV)) therefore remain a major challenge in diagnosing flavivirus infections. Virus neutralization tests are considered as reference tests for the detection of specific flavivirus antibodies, but are elaborate, time-consuming and need biosafety level 3 facilities. A simple and straightforward assay for the differentiation and detection of specific WNV IgG antibodies for the routine laboratory is urgently needed. In this study, we compared two commercially available enzyme-linked immunosorbent assays (anti-IgG WNV ELISA and anti-NS1-IgG WNV), a commercially available indirect immunofluorescence assay, and a newly developed in-house ELISA for the detection of WNV-NS1-IgG antibodies. All four tests were compared to an in-house NT to determine both the sensitivity and specificity of the four test systems. None of the assays could match the specificity of the NT, although the two NS1-IgG based ELISAs were very close to the specificity of the NT at 97.3% and 94.6%. The in-house WNV-NS1-IgG ELISA had the best performance regarding sensitivity and specificity. The specificities of the ELISA assays and the indirect immunofluorescence assays could not meet the necessary specificity and/or sensitivity.


Subject(s)
Antibodies, Viral , Enzyme-Linked Immunosorbent Assay , Sensitivity and Specificity , West Nile Fever , West Nile virus , West Nile virus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Humans , West Nile Fever/diagnosis , West Nile Fever/immunology , Enzyme-Linked Immunosorbent Assay/methods , Serologic Tests/methods , Immunoglobulin G/blood , Immunoglobulin G/immunology , Fluorescent Antibody Technique, Indirect/methods , Cross Reactions/immunology , Animals
6.
Front Immunol ; 15: 1343024, 2024.
Article in English | MEDLINE | ID: mdl-38784378

ABSTRACT

Background: Around 20% of the population in Northern and Central Europe is affected by birch pollen allergy, with the major birch pollen allergen Bet v 1 as the main elicitor of allergic reactions. Together with its cross-reactive allergens from related trees and foods, Bet v 1 causes an impaired quality of life. Hence, new treatment strategies were elaborated, demonstrating the effectiveness of blocking IgG antibodies on Bet v 1-induced IgE-mediated reactions. A recent study provided evidence for the first time that Bet v 1-specific nanobodies reduce patients´ IgE binding to Bet v 1. In order to increase the potential to outcompete IgE recognition of Bet v 1 and to foster cross-reactivity and cross-protection, we developed Bet v 1-specific nanobody trimers and evaluated their capacity to suppress polyclonal IgE binding to corresponding allergens and allergen-induced basophil degranulation. Methods: Nanobody trimers were engineered by adding isoleucine zippers, thus enabling trimeric formation. Trimers were analyzed for their cross-reactivity, binding kinetics to Bet v 1, and related allergens, and patients' IgE inhibition potential. Finally, their efficacy to prevent basophil degranulation was investigated. Results: Trimers showed enhanced recognition of cross-reactive allergens and increased efficiency to reduce IgE-allergen binding compared to nanobody monomers. Furthermore, trimers displayed slow dissociation rates from allergens and suppressed allergen-induced mediator release. Conclusion: We generated high-affine nanobody trimers that target Bet v 1 and related allergens. Trimers blocked IgE-allergen interaction by competing with IgE for allergen binding. They inhibited IgE-mediated release of biological mediators, demonstrating a promising potential to prevent allergic reactions caused by Bet v 1 and relatives.


Subject(s)
Allergens , Antigens, Plant , Cross Reactions , Immunoglobulin E , Single-Domain Antibodies , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Humans , Antigens, Plant/immunology , Single-Domain Antibodies/immunology , Cross Reactions/immunology , Allergens/immunology , Basophils/immunology , Basophils/metabolism , Protein Binding , Rhinitis, Allergic, Seasonal/immunology , Protein Multimerization
7.
Nat Commun ; 15(1): 4330, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773072

ABSTRACT

The Hendra and Nipah viruses (HNVs) are highly pathogenic pathogens without approved interventions for human use. In addition, the interaction pattern between the attachment (G) and fusion (F) glycoproteins required for virus entry remains unclear. Here, we isolate a panel of Macaca-derived G-specific antibodies that cross-neutralize HNVs via multiple mechanisms. The most potent antibody, 1E5, confers adequate protection against the Nipah virus challenge in female hamsters. Crystallography demonstrates that 1E5 has a highly similar binding pattern to the receptor. In cryo-electron microscopy studies, the tendency of 1E5 to bind to the upper or lower heads results in two distinct quaternary structures of G. Furthermore, we identify the extended outer loop ß1S2-ß1S3 of G and two pockets on the apical region of fusion (F) glycoprotein as the essential sites for G-F interactions. This work highlights promising drug candidates against HNVs and contributes deeper insights into the viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Cryoelectron Microscopy , Henipavirus Infections , Viral Fusion Proteins , Animals , Antibodies, Neutralizing/immunology , Female , Antibodies, Viral/immunology , Henipavirus Infections/virology , Henipavirus Infections/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/chemistry , Humans , Viral Envelope Proteins/immunology , Viral Envelope Proteins/chemistry , Nipah Virus/immunology , Virus Internalization/drug effects , Henipavirus/immunology , Cricetinae , Cross Reactions/immunology , Hendra Virus/immunology , Macaca , Mesocricetus , Crystallography, X-Ray
8.
Sci Rep ; 14(1): 11145, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750087

ABSTRACT

The global distribution of tropical fire ants (Solenopsis geminata) raises concerns about anaphylaxis and serious medical issues in numerous countries. This investigation focused on the cross-reactivity of allergen-specific IgE antibodies between S. geminata and Myrmecia pilosula (Jack Jumper ant) venom proteins due to the potential emergence of cross-reactive allergies in the future. Antibody epitope analysis unveiled one predominant conformational epitope on Sol g 1.1 (PI score of 0.989), followed by Sol g 2.2, Sol g 4.1, and Sol g 3.1. Additionally, Pilosulin 1 showed high allergenic potential (PI score of 0.94), with Pilosulin 5a (PI score of 0.797) leading in B-cell epitopes. The sequence analysis indicated that Sol g 2.2 and Sol g 4.1 pose a high risk of cross-reactivity with Pilosulins 4.1a and 5a. Furthermore, the cross-reactivity of recombinant Sol g proteins with M. pilosula-specific IgE antibodies from 41 patients revealed high cross-reactivity for r-Sol g 3.1 (58.53%) and r-Sol g 4.1 (43.90%), followed by r-Sol g 2.2 (26.82%), and r-Sol g 1.1 (9.75%). Therefore, this study demonstrates cross-reactivity (85.36%) between S. geminata and M. pilosula, highlighting the allergenic risk. Understanding these reactions is vital for the prevention of severe allergic reactions, especially in individuals with pre-existing Jumper Jack ant allergy, informing future management strategies.


Subject(s)
Allergens , Ant Venoms , Ants , Cross Reactions , Epitopes , Immunoglobulin E , Immunoglobulin E/immunology , Cross Reactions/immunology , Animals , Humans , Ant Venoms/immunology , Ants/immunology , Allergens/immunology , Epitopes/immunology , Recombinant Proteins/immunology , Insect Proteins/immunology , Female , Adult , Male , Amino Acid Sequence , Middle Aged , Adolescent , Young Adult
9.
Front Immunol ; 15: 1382911, 2024.
Article in English | MEDLINE | ID: mdl-38807606

ABSTRACT

Introduction: COVID-19 vaccines are highly effective in inducing protective immunity. While the serum antibody response to COVID-19 vaccination has been studied in depth, our knowledge of the underlying plasmablast and memory B cell (Bmem) responses is still incomplete. Here, we determined the antibody and B cell response to COVID-19 vaccination in a naïve population and contrasted it with the response to a single influenza vaccination in a primed cohort. In addition, we analyzed the antibody and B cell responses against the four endemic human coronaviruses (HCoVs). Methods: Measurement of specific plasma IgG antibodies was combined with functional analyses of antibody-secreting plasmablasts and Bmems. SARS-CoV-2- and HCoV-specific IgG antibodies were quantified with an in-house bead-based multiplexed immunoassay. Results: The antibody and B cell responses to COVID-19 vaccination reflected the kinetics of a prime-boost immunization, characterized by a slow and moderate primary response and a faster and stronger secondary response. In contrast, the influenza vaccinees possessed robust immune memory for the vaccine antigens prior to vaccination, and the recall vaccination moderately boosted antibody production and Bmem responses. Antibody levels and Bmem responses waned several months after the 2nd COVID-19 vaccination, but were restored upon the 3rd vaccination. The COVID-19 vaccine-induced antibodies mainly targeted novel, non-cross-reactive S1 epitopes of the viral spike protein, while cross-reactive S2 epitopes were less immunogenic. Booster vaccination not only strongly enhanced neutralizing antibodies against an original SARS-CoV-2 strain, but also induced neutralizing antibodies against the Omicron BA.2 variant. We observed a 100% plasma antibody prevalence against the S1 subunits of HCoVs, which was not affected by vaccination. Discussion: Overall, by complementing classical serology with a functional evaluation of plasmablasts and memory B cells we provide new insights into the specificity of COVID-19 vaccine-induced antibody and B cell responses.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Cross Reactions , Immunity, Humoral , Immunoglobulin G , Memory B Cells , Plasma Cells , SARS-CoV-2 , Humans , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , Memory B Cells/immunology , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , Male , Adult , Cross Reactions/immunology , Female , Plasma Cells/immunology , Middle Aged , Immunoglobulin G/immunology , Immunoglobulin G/blood , Vaccination , Influenza Vaccines/immunology , Immunologic Memory/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Epitopes, B-Lymphocyte/immunology , B-Lymphocytes/immunology , Spike Glycoprotein, Coronavirus/immunology , Kinetics
10.
Emerg Microbes Infect ; 13(1): 2356153, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38767199

ABSTRACT

Men who have sex with men and people living with HIV are disproportionately affected in the 2022 multi-country monkeypox epidemic. The smallpox vaccine can induce cross-reactive antibodies against the monkeypox virus (MPXV) and reduce the risk of infection. Data on antibodies against MPXV induced by historic smallpox vaccination in people with HIV are scarce. In this observational study, plasma samples were collected from people living with and without HIV in Shenzhen, China. We measured antibodies binding to two representative proteins of vaccinia virus (VACV; A27L and A33R) and homologous proteins of MPXV (A29L and A35R) using an enzyme-linked immunosorbent assay. We compared the levels of these antibodies between people living with and without HIV. Stratified analyses were performed based on the year of birth of 1981 when the smallpox vaccination was stopped in China. Plasma samples from 677 people living with HIV and 746 people without HIV were tested. A consistent pattern was identified among the four antibodies, regardless of HIV status. VACV antigen-reactive and MPXV antigen-reactive antibodies induced by historic smallpox vaccination were detectable in the people born before 1981, and antibody levels reached a nadir during or after 1981. The levels of smallpox vaccine-induced antibodies were comparable between people living with HIV and those without HIV. Our findings suggest that the antibody levels against MPXV decreased in both people living with and without HIV due to the cessation of smallpox vaccination.


Subject(s)
Antibodies, Viral , HIV Infections , Monkeypox virus , Smallpox Vaccine , Humans , Antibodies, Viral/blood , Antibodies, Viral/immunology , Male , Smallpox Vaccine/immunology , Smallpox Vaccine/administration & dosage , HIV Infections/immunology , HIV Infections/epidemiology , HIV Infections/virology , Adult , Female , China/epidemiology , Middle Aged , Monkeypox virus/immunology , Smallpox/immunology , Smallpox/prevention & control , Smallpox/epidemiology , Smallpox/history , Vaccination , Mpox (monkeypox)/immunology , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/history , Cross Reactions/immunology , Young Adult , Enzyme-Linked Immunosorbent Assay , Vaccinia virus/immunology
11.
BMC Immunol ; 25(1): 29, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38730320

ABSTRACT

BACKGROUND: Several PD-1 antibodies approved as anti-cancer therapies work by blocking the interaction of PD-1 with its ligand PD-L1, thus restoring anti-cancer T cell activities. These PD-1 antibodies lack inter-species cross-reactivity, necessitating surrogate antibodies for preclinical studies, which may limit the predictability and translatability of the studies. RESULTS: To overcome this limitation, we have developed an inter-species cross-reactive PD-1 antibody, GNUV201, by utilizing an enhanced diversity mouse platform (SHINE MOUSE™). GNUV201 equally binds to human PD-1 and mouse PD-1, equally inhibits the binding of human PD-1/PD-L1 and mouse PD-1/PD-L1, and effectively suppresses tumor growth in syngeneic mouse models. The epitope of GNUV201 mapped to the "FG loop" of hPD-1, distinct from those of Keytruda® ("C'D loop") and Opdivo® (N-term). Notably, the structural feature where the protruding epitope loop fits into GNUV201's binding pocket supports the enhanced binding affinity due to slower dissociation (8.7 times slower than Keytruda®). Furthermore, GNUV201 shows a stronger binding affinity at pH 6.0 (5.6 times strong than at pH 7.4), which mimics the hypoxic and acidic tumor microenvironment (TME). This phenomenon is not observed with marketed antibodies (Keytruda®, Opdivo®), implying that GNUV201 achieves more selective binding to and better occupancy on PD-1 in the TME. CONCLUSIONS: In summary, GNUV201 exhibited enhanced affinity for PD-1 with slow dissociation and preferential binding in TME-mimicking low pH. Human/monkey/mouse inter-species cross-reactivity of GNUV201 could enable more predictable and translatable efficacy and toxicity preclinical studies. These results suggest that GNUV201 could be an ideal antibody candidate for anti-cancer drug development.


Subject(s)
Cross Reactions , Immunotherapy , Programmed Cell Death 1 Receptor , Animals , Humans , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Mice , Cross Reactions/immunology , Immunotherapy/methods , Hydrogen-Ion Concentration , Neoplasms/immunology , Neoplasms/therapy , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , Cell Line, Tumor , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Epitopes/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Mice, Inbred C57BL , Female
12.
PLoS One ; 19(5): e0302722, 2024.
Article in English | MEDLINE | ID: mdl-38722827

ABSTRACT

BACKGROUND: Pakistan is endemic to a diverse set of parasitic, mycobacterial and viral diseases. The recognition of BCG Trained Immunity (TI) led us to postulate that the continued presence of BCG-TI may play a protective role, previously reported for both infectious and noninfectious conditions. Most of the previous studies have addressed the issue of BCG-TI in the paediatric populations. This study addressed the key issue of maintenance of BCG-TI in a wider age range (adolescent and adults) to identify the strength and quality of the immune responses. OBJECTIVE: To assess the BCG-induced recall responses in healthy individuals by cytokines secreted from the TI network and its potential role in providing cross-protection against COVID-19 and other viral infections. STUDY DESIGN: In this cross-sectional study, healthy young adults and adolescents (n = 20) were recruited from 16-40 years of age, with no prior history of TB treatment, autoimmune, or chronic inflammatory condition. METHODS: BCG-induced cytokine responses were assessed using prototypic markers for cells of the TI network [macrophages [M1 (TNFα, IFNγ), M2 (IL10)], NK (IL2), Gamma delta (γδ) T (IL17, IL4)] and SARS CoV2 IgG antibodies against RBD using short-term (12 hrs.) cultures assay. RESULTS: Significant differences were observed in the magnitude of recall responses to BCG with macrophage cytokines showing the highest mean levels of TNFα (9148 pg/ml) followed by IL10 (488 pg/ml) and IFNγ (355 pg/ml). The ratio of unstimulated vs.BCG-stimulated cytokines was 132 fold higher for TNFα, 40 fold fo r IL10, and 27 fold for IFNγ. Furthermore, SARS-CoV-2 antibodies were also detected in unstimulated plasma which showed cross reactivity with BCG. CONCLUSION: The presence of cross reactive antibodies to SARS-CoV-2 and the relative ratio of pro- and anti-inflammatory cytokines secreted by activated TI cellular network may play a pivotal role in protection in the early stages of infection as observed during the COVID-19 pandemic in the younger age groups resulting in lower morbidity and mortality.


Subject(s)
Antibodies, Viral , BCG Vaccine , COVID-19 , Cytokines , SARS-CoV-2 , Humans , BCG Vaccine/immunology , Adult , COVID-19/immunology , COVID-19/prevention & control , Adolescent , Cross-Sectional Studies , Male , Female , SARS-CoV-2/immunology , Cytokines/immunology , Young Adult , Antibodies, Viral/immunology , Antibodies, Viral/blood , Cross Reactions/immunology , Vaccination , Pakistan/epidemiology , Trained Immunity
13.
PLoS Pathog ; 20(4): e1012159, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38662650

ABSTRACT

Human enteroviruses are the most common human pathogen with over 300 distinct genotypes. Previous work with poliovirus has suggested that it is possible to generate antibody responses in humans and animals that can recognize members of multiple enterovirus species. However, cross protective immunity across multiple enteroviruses is not observed epidemiologically in humans. Here we investigated whether immunization of mice or baboons with inactivated poliovirus or enterovirus virus-like-particles (VLPs) vaccines generates antibody responses that can recognize enterovirus D68 or A71. We found that mice only generated antibodies specific for the antigen they were immunized with, and repeated immunization failed to generate cross-reactive antibody responses as measured by both ELISA and neutralization assay. Immunization of baboons with IPV failed to generate neutralizing antibody responses against enterovirus D68 or A71. These results suggest that a multivalent approach to enterovirus vaccination is necessary to protect against enterovirus disease in vulnerable populations.


Subject(s)
Antibodies, Viral , Cross Reactions , Enterovirus Infections , Poliovirus Vaccine, Inactivated , Animals , Mice , Cross Reactions/immunology , Antibodies, Viral/immunology , Enterovirus Infections/immunology , Enterovirus Infections/prevention & control , Enterovirus Infections/virology , Poliovirus Vaccine, Inactivated/immunology , Poliovirus Vaccine, Inactivated/administration & dosage , Vaccines, Virus-Like Particle/immunology , Antibodies, Neutralizing/immunology , Papio/immunology , Humans , Poliovirus/immunology , Female , Antibody Formation/immunology , Enterovirus/immunology , Mice, Inbred BALB C , Enterovirus D, Human/immunology
14.
Brief Bioinform ; 25(3)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38653491

ABSTRACT

Coronaviruses have threatened humans repeatedly, especially COVID-19 caused by SARS-CoV-2, which has posed a substantial threat to global public health. SARS-CoV-2 continuously evolves through random mutation, resulting in a significant decrease in the efficacy of existing vaccines and neutralizing antibody drugs. It is critical to assess immune escape caused by viral mutations and develop broad-spectrum vaccines and neutralizing antibodies targeting conserved epitopes. Thus, we constructed CovEpiAb, a comprehensive database and analysis resource of human coronavirus (HCoVs) immune epitopes and antibodies. CovEpiAb contains information on over 60 000 experimentally validated epitopes and over 12 000 antibodies for HCoVs and SARS-CoV-2 variants. The database is unique in (1) classifying and annotating cross-reactive epitopes from different viruses and variants; (2) providing molecular and experimental interaction profiles of antibodies, including structure-based binding sites and around 70 000 data on binding affinity and neutralizing activity; (3) providing virological characteristics of current and past circulating SARS-CoV-2 variants and in vitro activity of various therapeutics; and (4) offering site-level annotations of key functional features, including antibody binding, immunological epitopes, SARS-CoV-2 mutations and conservation across HCoVs. In addition, we developed an integrated pipeline for epitope prediction named COVEP, which is available from the webpage of CovEpiAb. CovEpiAb is freely accessible at https://pgx.zju.edu.cn/covepiab/.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Epitopes , SARS-CoV-2 , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Antibodies, Neutralizing/immunology , Epitopes/immunology , Epitopes/chemistry , Epitopes/genetics , Coronavirus/immunology , Coronavirus/genetics , Databases, Factual , Cross Reactions/immunology
15.
Nat Commun ; 15(1): 3387, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684663

ABSTRACT

Influenza B viruses (IBVs) cause substantive morbidity and mortality, and yet immunity towards IBVs remains understudied. CD8+ T-cells provide broadly cross-reactive immunity and alleviate disease severity by recognizing conserved epitopes. Despite the IBV burden, only 18 IBV-specific T-cell epitopes restricted by 5 HLAs have been identified currently. A broader array of conserved IBV T-cell epitopes is needed to develop effective cross-reactive T-cell based IBV vaccines. Here we identify 9 highly conserved IBV CD8+ T-cell epitopes restricted to HLA-B*07:02, HLA-B*08:01 and HLA-B*35:01. Memory IBV-specific tetramer+CD8+ T-cells are present within blood and tissues. Frequencies of IBV-specific CD8+ T-cells decline with age, but maintain a central memory phenotype. HLA-B*07:02 and HLA-B*08:01-restricted NP30-38 epitope-specific T-cells have distinct T-cell receptor repertoires. We provide structural basis for the IBV HLA-B*07:02-restricted NS1196-206 (11-mer) and HLA-B*07:02-restricted NP30-38 epitope presentation. Our study increases the number of IBV CD8+ T-cell epitopes, and defines IBV-specific CD8+ T-cells at cellular and molecular levels, across tissues and age.


Subject(s)
CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Influenza B virus , Influenza, Human , CD8-Positive T-Lymphocytes/immunology , Humans , Epitopes, T-Lymphocyte/immunology , Influenza B virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Adult , Middle Aged , Aged , Cross Reactions/immunology , Young Adult , Female , Male , Immunologic Memory/immunology , Adolescent , HLA-B Antigens/immunology , Child , Child, Preschool
16.
Viruses ; 16(4)2024 04 17.
Article in English | MEDLINE | ID: mdl-38675961

ABSTRACT

AIMS: To evaluate whether antibodies specific for the vaccinia virus (VV) are still detectable after at least 45 years from immunization. To confirm that VV-specific antibodies are endowed with the capacity to neutralize Mpox virus (MPXV) in vitro. To test a possible role of polyclonal non-specific activation in the maintenance of immunologic memory. METHODS: Sera were collected from the following groups: smallpox-vaccinated individuals with or without latent tuberculosis infection (LTBI), unvaccinated donors, and convalescent individuals after MPXV infection. Supernatant of VV- or MPXV-infected Vero cells were inactivated and used as antigens in ELISA or in Western blot (WB) analyses. An MPXV plaque reduction neutralization test (PRNT) was optimized and performed on study samples. VV- and PPD-specific memory T cells were measured by flow cytometry. RESULTS: None of the smallpox unvaccinated donors tested positive in ELISA or WB analysis and their sera were unable to neutralize MPXV in vitro. Sera from all the individuals convalescing from an MPXV infection tested positive for anti-VV or MPXV IgG with high titers and showed MPXV in vitro neutralization capacity. Sera from most of the vaccinated individuals showed IgG anti-VV and anti-MPXV at high titers. WB analyses showed that positive sera from vaccinated or convalescent individuals recognized both VV and MPXV antigens. Higher VV-specific IgG titer and specific T cells were observed in LTBI individuals. CONCLUSIONS: ELISA and WB performed using supernatant of VV- or MPXV-infected cells are suitable to identify individuals vaccinated against smallpox at more than 45 years from immunization and individuals convalescing from a recent MPXV infection. ELISA and WB results show a good correlation with PRNT. Data confirm that a smallpox vaccination induces a long-lasting memory in terms of specific IgG and that antibodies raised against VV may neutralize MPXV in vitro. Finally, higher titers of VV-specific antibodies and higher frequency of VV-specific memory T cells in LTBI individuals suggest a role of polyclonal non-specific activation in the maintenance of immunologic memory.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , B-Lymphocytes , Cross Reactions , Smallpox Vaccine , Vaccinia virus , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Smallpox Vaccine/immunology , B-Lymphocytes/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Cross Reactions/immunology , Vaccinia virus/immunology , Middle Aged , Immunologic Memory , Neutralization Tests , Smallpox/immunology , Smallpox/prevention & control , Animals , Male , T-Lymphocytes/immunology , Female , Enzyme-Linked Immunosorbent Assay , Orthopoxvirus/immunology , Vaccination , Chlorocebus aethiops , Adult , Lymphocyte Activation , Vero Cells
17.
Expert Rev Vaccines ; 23(1): 474-484, 2024.
Article in English | MEDLINE | ID: mdl-38632930

ABSTRACT

INTRODUCTION: Anti-neuraminidase (NA) immunity correlates with the protection against influenza virus infection in both human and animal models. The aim of this review is to better understand the mechanism of anti-NA immunity, and also to evaluate the approaches on developing NA-based influenza vaccines or enhancing immune responses against NA for current influenza vaccines. AREAS COVERED: In this review, the structure of influenza neuraminidase, the contribution of anti-NA immunity to protection, as well as the efforts and challenges of targeting the immune responses to NA were discussed. We also listed some of the newly discovered anti-NA monoclonal antibodies and discussed their contribution in therapeutic as well as the antigen design of a broadly protective NA vaccine. EXPERT OPINION: Targeting the immune response to both HA and NA may be critical for achieving the optimal protection since there are different mechanisms of HA and NA elicited protective immunity. Monoclonal antibodies (mAbs) that target the conserved protective lateral face or catalytic sites are effective therapeutics. The epitope discovery using monoclonal antibodies may benefit NA-based vaccine elicited broadly reactive antibody responses. Therefore, the potential for a vaccine that elicits cross-reactive antibodies against neuraminidase is a high priority for next-generation influenza vaccines.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Influenza Vaccines , Influenza, Human , Neuraminidase , Humans , Neuraminidase/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Monoclonal/immunology , Animals , Antibodies, Viral/immunology , Vaccine Development , Cross Reactions/immunology , Epitopes/immunology
18.
Vaccine ; 42(15): 3445-3454, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38631956

ABSTRACT

Major histocompatibility complex class II (MHC-II) molecules are involved in immune responses against pathogens and vaccine candidates' immunogenicity. Immunopeptidomics for identifying cancer and infection-related antigens and epitopes have benefited from advances in immunopurification methods and mass spectrometry analysis. The mouse anti-MHC-II-DR monoclonal antibody L243 (mAb-L243) has been effective in recognising MHC-II-DR in both human and non-human primates. It has also been shown to cross-react with other animal species, although it has not been tested in livestock. This study used mAb-L243 to identify Staphylococcus aureus and Salmonella enterica serovar Typhimurium peptides binding to cattle and swine macrophage MHC-II-DR molecules using flow cytometry, mass spectrometry and two immunopurification techniques. Antibody cross-reactivity led to identifying expressed MHC-II-DR molecules, together with 10 Staphylococcus aureus peptides in cattle and 13 S. enterica serovar Typhimurium peptides in swine. Such data demonstrates that MHC-II-DR expression and immunocapture approaches using L243 mAb represents a viable strategy for flow cytometry and immunopeptidomics analysis of bovine and swine antigen-presenting cells.


Subject(s)
Antibodies, Monoclonal , Macrophages , Salmonella typhimurium , Staphylococcus aureus , Animals , Cattle , Swine/immunology , Staphylococcus aureus/immunology , Antibodies, Monoclonal/immunology , Macrophages/immunology , Salmonella typhimurium/immunology , Histocompatibility Antigens Class II/immunology , Cross Reactions/immunology , Flow Cytometry , Mass Spectrometry , Mice
19.
Biol Chem ; 405(6): 367-381, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38662449

ABSTRACT

Structural and allergenic characterization of mite profilins has not been previously pursued to a similar extent as plant profilins. Here, we describe structures of profilins originating from Tyrophagus putrescentiae (registered allergen Tyr p 36.0101) and Dermatophagoides pteronyssinus (here termed Der p profilin), which are the first structures of profilins from Arachnida. Additionally, the thermal stabilities of mite and plant profilins are compared, suggesting that the high number of cysteine residues in mite profilins may play a role in their increased stability. We also examine the cross-reactivity of plant and mite profilins as well as investigate the relevance of these profilins in mite inhalant allergy. Despite their high structural similarity to other profilins, mite profilins have low sequence identity with plant and human profilins. Subsequently, these mite profilins most likely do not display cross-reactivity with plant profilins. At the same time the profilins have highly conserved poly(l-proline) and actin binding sites.


Subject(s)
Cross Reactions , Profilins , Animals , Cross Reactions/immunology , Profilins/immunology , Profilins/chemistry , Profilins/metabolism , Humans , Mites/immunology , Mites/chemistry , Amino Acid Sequence , Hypersensitivity/immunology , Plants/immunology , Plants/chemistry , Plants/metabolism , Models, Molecular , Allergens/immunology , Allergens/chemistry
20.
Mol Immunol ; 170: 19-25, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38598870

ABSTRACT

The assembly of tissue-damaging membrane attack complexes (MACs; C5b-9) is a major mechanism by which excessive complement activation causes diseases. We previously developed a mouse anti-human C6 monoclonal antibody (mAb) 1C9 that selectively inhibits the assembly of MACs in human and non-human primates. In this project, we found that 1C9 also cross-reacted with rat and guinea pig C6, and determined its binding domains on C6 using different truncated C6 proteins. We then humanized the anti-C6 mAb by molecular modeling and complementarity-determining region grafting. After screening a library of 276 humanized variants with different combinations of humanized light and heavy chains in biophysical assays, we identified clone 3713 with the best developability profile, and an increased affinity against C6 when compared with the parental 1C9 mAb. This humanized 3713 mAb inhibited human, monkey, and rat complement-mediated hemolysis in vitro, and more importantly, it significantly reduced complement-mediated hemolysis in vivo in rats. These results demonstrated the successful humanization of the anti-C6 mAb and suggested that the humanized 3713 mAb could be further developed as a new therapeutic that selectively targets MAC for certain complement-mediated pathological conditions.


Subject(s)
Antibodies, Monoclonal , Complement C6 , Hemolysis , Animals , Humans , Rats , Guinea Pigs , Mice , Hemolysis/drug effects , Hemolysis/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Complement C6/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/immunology , Complement Activation/drug effects , Complement Membrane Attack Complex/immunology , Cross Reactions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...