Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62.256
Filter
1.
J Transl Med ; 22(1): 827, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242525

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are pivotal in combating coronavirus disease 2019 (COVID-19); however, the declining antibody titers postvaccination pose challenges for sustained protection and herd immunity. Although gut microbiome is reported to affect the early antibody response after vaccination, its impact on the longevity of vaccine-induced antibodies remains unexplored. METHODS: A prospective cohort study was conducted involving 44 healthy adults who received two doses of either the BNT162b2 or ChAdOx1 vaccine, followed by a BNT162b2 booster at six months. The gut microbiome was serially analyzed using 16S rRNA and shotgun sequencing, while humoral immune response was assessed using a SARS-CoV-2 spike protein immunoassay. RESULTS: Faecalibacterium prausnitzii was associated with robust and persistent antibody responses post-BNT162b2 vaccination. In comparison, Escherichia coli was associated with a slower antibody decay following ChAdOx1 vaccination. The booster immune response was correlated with metabolic pathways involving cellular functions and aromatic amino acid synthesis. CONCLUSIONS: The findings of this study underscored the potential interaction between the gut microbiome and the longevity/boosting effect of antibodies following vaccination against SARS-CoV-2. The identification of specific microbial associations suggests the prospect of microbiome-based strategies for enhancing vaccine efficacy.


Subject(s)
Antibodies, Viral , BNT162 Vaccine , COVID-19 , Gastrointestinal Microbiome , Immunization, Secondary , SARS-CoV-2 , Vaccination , Humans , Gastrointestinal Microbiome/immunology , Male , Female , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , BNT162 Vaccine/immunology , COVID-19 Vaccines/immunology , Middle Aged , ChAdOx1 nCoV-19/immunology , Prospective Studies , Antibody Formation/immunology , Spike Glycoprotein, Coronavirus/immunology , Immunity, Humoral/immunology , Young Adult
2.
BMC Gastroenterol ; 24(1): 329, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350092

ABSTRACT

BACKGROUND: Metabolic dysfunction-associated steatotic liver disease (MASLD) patients are at an elevated risk of developing severe coronavirus disease 2019 (COVID-19). The objective of this study was to assess antibody responses and safety profiles six months after the third dose of the inactivated acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine in MASLD patients. METHODS: This study included MASLD patients and healthy volunteers without a history of SARS-CoV-2 infection. Blood samples were collected six months after receiving the third dose of the inactivated vaccine to measure the levels of neutralizing antibodies (NAbs) and anti-spike IgG antibodies against SARS-CoV-2. RESULTS: A total of 335 participants (214 MASLD patients and 121 healthy volunteers) were enrolled. The seroprevalence of NAb was 61.7% (132 of 214) in MASLD patients and 74.4% (90 of 121) in healthy volunteers, which was a significant difference (p = 0.018). Statistically significant differences in IgG seroprevalence were also observed between MASLD patients and healthy volunteers (p = 0.004). Multivariate analysis demonstrated that the severity of MASLD (OR, 2.97; 95% CI, 1.32-6.68; p = 0.009) and age (OR, 1.03; 95% CI, 1.01-1.06; p = 0.004) were independent risk factors for NAb negativity in MASLD patients. Moderate/severe MASLD patients had a lower NAb seroprevalence than mild MASLD patients (45.0% vs. 65.5%, p = 0.016). CONCLUSION: Lower antibody responses were observed in MASLD patients six months after their third dose of the inactivated vaccine than in healthy volunteers, providing further assistance in monitoring patients who are more vulnerable to hypo-responsiveness to SARS-CoV-2 vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunoglobulin G , SARS-CoV-2 , Vaccines, Inactivated , Humans , Male , Female , Middle Aged , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Adult , SARS-CoV-2/immunology , Immunoglobulin G/blood , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Aged , Antibody Formation/immunology , Seroepidemiologic Studies
3.
PLoS Biol ; 22(9): e3002800, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39316608

ABSTRACT

Bats are reservoirs of many zoonotic viruses that are fatal in humans but do not cause disease in bats. Moreover, bats generate low neutralizing antibody titers in response to experimental viral infection, although more robust antibody responses have been observed in wild-caught bats during times of food stress. Here, we compared the antibody titers and B cell receptor (BCR) diversity of Jamaican fruit bats (Artibeus jamaicensis; JFBs) and BALB/c mice generated in response to T-dependent and T-independent antigens. We then manipulated the diet of JFBs and challenged them with H18N11 influenza A-like virus or a replication incompetent Nipah virus VSV (Nipah-riVSV). Under standard housing conditions, JFBs generated a lower avidity antibody response and possessed more BCR mRNA diversity compared to BALB/c mice. However, withholding protein from JFBs improved serum neutralization in response to Nipah-riVSV and improved serum antibody titers specific to H18 but reduced BCR mRNA diversity.


Subject(s)
Antibodies, Viral , Chiroptera , Mice, Inbred BALB C , Animals , Chiroptera/immunology , Chiroptera/virology , Mice , Antibodies, Viral/immunology , Nipah Virus/immunology , Antibody Formation/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Diet, Protein-Restricted , Antibodies, Neutralizing/immunology , Antibody Affinity , Influenza A virus/immunology , Female , Antibody Diversity
4.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337303

ABSTRACT

There is a need to investigate novel strategies in order to create an effective, broadly protective vaccine for current and future severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreaks. The currently available vaccines demonstrate compromised efficacy against emerging SARS-CoV-2 variants of concern (VOCs), short-lived immunity, and susceptibility to immune imprinting due to frequent boosting practices. In this study, we examined the specificity of cross-reactive IgG antibody responses in mRNA-vaccinated, AstraZeneca-vaccinated, and unvaccinated donors to identify potentially conserved, cross-reactive epitopes to target in order to create a broadly protective SARS-CoV-2 vaccine. Our study provides evidence for cross-reactive IgG antibodies specific to eight different spike (S) variants. Furthermore, the specificities of these cross-variant IgG antibody titers were associated to some extent with spike S1- and S2-subunit-derived epitopes P1 and P2, respectively. In addition, nucleocapsid (N)- and membrane (M)-specific IgG antibody titers correlated with N- and M-derived epitopes conserved across beta-CoVs, P3-7. This study reveals conserved epitopes of viral antigens, targeted by natural and/or vaccine-induced human immunity, for future designs of next-generation COVID-19 vaccines.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Cross Reactions , Epitopes , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Immunoglobulin G/immunology , Immunoglobulin G/blood , COVID-19 Vaccines/immunology , Cross Reactions/immunology , Epitopes/immunology , Female , Adult , Male , Middle Aged , Vaccination , Antibody Formation/immunology , Antigens, Viral/immunology
5.
Nat Microbiol ; 9(10): 2488-2505, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232205

ABSTRACT

Parasitic nematodes have an intimate, chronic and lifelong exposure to vertebrate tissues. Here we mined 41 published parasitic nematode transcriptomes from vertebrate hosts and identified 91 RNA viruses across 13 virus orders from 24 families in ~70% (28 out of 41) of parasitic nematode species, which include only 5 previously reported viruses. We observe widespread distribution of virus-nematode associations across multiple continents, suggesting an ancestral acquisition event and host-virus co-evolution. Characterization of viruses of Brugia malayi (BMRV1) and Onchocerca volvulus (OVRV1) shows that these viruses are abundant in reproductive tissues of adult parasites. Importantly, the presence of BMRV1 RNA in B. malayi parasites mounts an RNA interference response against BMRV1 suggesting active viral replication. Finally, BMRV1 and OVRV1 were found to elicit antibody responses in serum samples from infected jirds and infected or exposed humans, indicating direct exposure to the immune system.


Subject(s)
Brugia malayi , RNA Viruses , Animals , RNA Viruses/immunology , RNA Viruses/genetics , Humans , Brugia malayi/immunology , Brugia malayi/genetics , Onchocerca volvulus/immunology , Onchocerca volvulus/genetics , Vertebrates/virology , Vertebrates/immunology , Vertebrates/parasitology , Nematoda/immunology , Nematoda/genetics , Nematoda/virology , Transcriptome , Antibody Formation/immunology , Phylogeny , RNA Interference
6.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39337675

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease, mediated by a complex interaction between B cells and various subsets of T cells. Dysfunction of helper T (Th) and regulatory T (Treg) cells may contribute to the breakdown of self-tolerance and the progression of autoimmune disease. In this study, we investigated the activity of Th and Treg cells on the differentiation of autologous B cells in vitro using cell cultures from the peripheral blood of healthy controls (HCs) and RA patients. The expressions of programmed death 1 (PD-1) and IL-21 were monitored as activation markers for Th cells. Unstimulated Th cells from RA patients showed remarkably higher PD-1 expression than HC samples. Stimulation of Th cells from RA patients with Staphylococcus enterotoxin B (SEB) in the presence of B cells significantly induced their PD-1 and IL-21 expression at a considerably higher level in RA compared to HCs, and Treg cells did not affect IL-21 production. When monitoring B-cell differentiation, a significantly higher frequency of plasma cells was observed, even in unstimulated samples of RA patients compared to HCs. In the SEB-stimulated co-cultures of the RA samples, plasma cell frequency and IgG production were considerably higher than in HCs and were not significantly affected by Tregs. These findings demonstrate that Th cells are constitutively active in RA, and their hyperactivity upon interaction with diseased B cells may lead to uncontrolled antibody production.


Subject(s)
Arthritis, Rheumatoid , B-Lymphocytes , Interleukins , Programmed Cell Death 1 Receptor , T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Regulatory , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Female , Programmed Cell Death 1 Receptor/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Male , Interleukins/metabolism , Middle Aged , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antibody Formation/immunology , Cell Differentiation/immunology , Adult , Enterotoxins/immunology , Cells, Cultured , Aged , Lymphocyte Activation/immunology , Coculture Techniques
7.
Cell Rep ; 43(9): 114708, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39243373

ABSTRACT

Lassa fever continues to be a major public health burden in West Africa, yet effective therapies or vaccines are lacking. The isolation of protective neutralizing antibodies against the Lassa virus glycoprotein complex (GPC) justifies the development of vaccines that can elicit strong neutralizing antibody responses. However, Lassa vaccine candidates have generally been unsuccessful at doing so, and the associated antibody responses to these vaccines remain poorly characterized. Here, we establish an electron microscopy-based epitope mapping workflow that enables high-resolution structural characterization of polyclonal antibodies to the GPC. By applying this method to rabbits vaccinated with a recombinant GPC vaccine and a GPC-derived virus-like particle, we reveal determinants of neutralization that involve epitopes of the GPC-A competition cluster. Furthermore, by identifying undescribed immunogenic off-target epitopes, we expose the challenges that recombinant GPC vaccines face. By enabling detailed polyclonal antibody characterization, our work ushers in a next generation of more rational Lassa vaccine design.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Lassa Fever , Lassa virus , Lassa virus/immunology , Animals , Antibodies, Neutralizing/immunology , Rabbits , Antibodies, Viral/immunology , Lassa Fever/immunology , Lassa Fever/prevention & control , Epitopes/immunology , Viral Vaccines/immunology , Humans , Epitope Mapping , Antibody Formation/immunology
8.
Nat Immunol ; 25(10): 1809-1819, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39261722

ABSTRACT

Evolutionary pressures sculpt population genetics, whereas immune adaptation fortifies humans against life-threatening organisms. How the evolution of selective genetic variation in adaptive immune receptors orchestrates the adaptation of human populations to contextual perturbations remains elusive. Here, we show that the G396R coding variant within the human immunoglobulin G1 (IgG1) heavy chain presents a concentrated prevalence in Southeast Asian populations. We uncovered a 190-kb genomic linkage disequilibrium block peaked in close proximity to this variant, suggestive of potential Darwinian selection. This variant confers heightened immune resilience against various pathogens and viper toxins in mice. Mechanistic studies involving severe acute respiratory syndrome coronavirus 2 infection and vaccinated individuals reveal that this variant enhances pathogen-specific IgG1+ memory B cell activation and antibody production. This G396R variant may have arisen on a Neanderthal haplotype background. These findings underscore the importance of an IGHG1 variant in reinforcing IgG1 antibody responses against life-threatening organisms, unraveling the intricate interplay between human evolution and immune adaptation.


Subject(s)
COVID-19 , Immunoglobulin G , Immunoglobulin Heavy Chains , SARS-CoV-2 , Humans , Animals , Immunoglobulin G/immunology , COVID-19/immunology , COVID-19/genetics , SARS-CoV-2/immunology , Mice , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Linkage Disequilibrium , Antibody Formation/genetics , Antibody Formation/immunology , Haplotypes , Memory B Cells/immunology , Female , Genetic Variation , Male
9.
Cells ; 13(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273052

ABSTRACT

The complex structure of monoclonal antibodies (mAbs) expressed in Chinese hamster ovary (CHO) cells may result in the accumulation of unfolded proteins, triggering endoplasmic reticulum (ER) stress and an unfolded protein response (UPR). If the protein folding ability cannot maintain ER homeostasis, the cell will shut down protein translation and ultimately induce apoptosis. We co-overexpressed HsQSOX1b and survivin proteins in the antibody-producing cell line CHO-PAb to obtain a new cell line, CHO-PAb-QS. Compared with CHO-PAb cells, the survival time of CHO-PAb-QS cells in batch culture was extended by 2 days, and the antibody accumulation and productivity were increased by 52% and 45%, respectively. The proportion of (HC-LC)2 was approximately doubled in the CHO-PAb-QS cells, which adapted to the accelerated disulfide bond folding capacity by upregulating the UPR's strength and increasing the ER content. The results of the apoptosis assays indicated that the CHO-PAb-QS cell line exhibited more excellent resistance to apoptosis induced by ER stress. Finally, CHO-PAb-QS cells exhibited mild oxidative stress but did not significantly alter the redox status. This study demonstrated that strategies based on HsQSOX1b and survivin co-overexpression could facilitate protein disulfide bond folding and anti-apoptosis ability, enhancing antibody production efficiency in CHO cell lines.


Subject(s)
Apoptosis , Cricetulus , Disulfides , Protein Folding , CHO Cells , Animals , Disulfides/metabolism , Disulfides/chemistry , Endoplasmic Reticulum Stress , Unfolded Protein Response , Antibody Formation , Antibodies, Monoclonal , Cricetinae , Survivin/metabolism , Humans , Endoplasmic Reticulum/metabolism , Oxidative Stress
10.
J Med Virol ; 96(9): e29862, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39247972

ABSTRACT

Limited research suggests that certain viruses reactivate in severe-acute-respiratory-syndrome-coronavirus 2 infection, contributing to the development of postacute sequelae of COVID-19 (PASC). We examined 1083 infected individuals from a population-based cohort, and assessed differences in plasma immunoglobulin (Ig)G and immunoglobulin A levels against Epstein-Barr virus (EBV), cytomegalovirus, varicella zoster virus (VZV), BK polyomavirus, KI polyomavirus, WU polyomavirus (WUPyV), respiratory syncytial virus, and Adv-36 according to the severity of previous COVID-19 and PASC history. Individuals who had experienced severe COVID-19 had higher antibody responses to latent viruses. Ever PASC, active persistent PASC, and PASC with neuropsychiatric symptoms were associated with higher immnoglobulin G to EBV early antigen-diffuse, VZV, and WUPyV even among individuals without previous severe COVID-19.


Subject(s)
Antibodies, Viral , COVID-19 , Immunoglobulin G , Humans , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/blood , Male , Female , Middle Aged , Immunoglobulin G/blood , Adult , Severity of Illness Index , Aged , SARS-CoV-2/immunology , Immunoglobulin A/blood , Antibody Formation , Post-Acute COVID-19 Syndrome , Cohort Studies
11.
Cytokine ; 183: 156748, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39241273

ABSTRACT

Enhanced IgG4 antibody (Ab) response is a prominent feature of type 1 autoimmune pancreatitis (AIP). Innate immune responses associated with IgG4 Ab production are poorly defined. We have previously reported that peripheral blood mononuclear cells (PBMCs) isolated from patients with type 1 AIP produce large amounts of IgG4 Abs upon stimulation with bacterial cell wall components. In addition, we showed that activation of plasmacytoid dendritic cells producing interferon (IFN)-α, interleukin (IL)-33, and B cell-activating factor (BAFF) upon sensing intestinal bacteria mediates the development of experimental AIP. In this study, we attempted to clarify the role of innate immunity against fungi in inducing enhanced IgG4 Ab responses in type 1 AIP. PBMCs isolated from healthy controls and patients with type 1 AIP were stimulated with a broad range of bacterial and fungal cell wall components. The concentrations of IgG1, IgG4, and cytokines were measured using enzyme-linked immunosorbent assays. Cell wall components derived from bacteria and fungi induced IgG1 and IgG4 Ab production in patients with type 1 AIP. Various types of microbe-associated molecular pattern motifs enhanced IgG4 Ab production in patients with type 1 AIP compared with the limited motifs in healthy controls. The enhanced IgG1 and IgG4 Ab production that followed in response to bacterial and fungal cell wall components was parallel to that of IFN-α, IFN-γ, IL-10, IL-33, and BAFF. In conclusion, cell wall components derived from fungi as well as bacteria promote IgG4 Ab responses in patients with type 1 AIP.


Subject(s)
Autoimmune Pancreatitis , Fungi , Immunoglobulin G , Leukocytes, Mononuclear , Humans , Immunoglobulin G/immunology , Male , Female , Middle Aged , Autoimmune Pancreatitis/immunology , Autoimmune Pancreatitis/microbiology , Fungi/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Aged , Bacteria/immunology , Cell Wall/immunology , Cell Wall/metabolism , Cytokines/metabolism , Cytokines/immunology , Adult , Antibody Formation/immunology , Immunity, Innate/immunology
12.
Vaccine ; 42(25): 126206, 2024 Nov 14.
Article in English | MEDLINE | ID: mdl-39122634

ABSTRACT

BACKGROUND: Kidney transplant recipients (KTRs) show poor antibody response to the SARS-CoV-2 vaccine. There is limited data on immune response to non-mRNA vaccines in KTRs. We studied the antibody response to the SARS-CoV-2 non-mRNA vaccine in a cohort of kidney transplant recipients. METHODS: We included KTRs following up in the tertiary care transplant outpatient clinic from February to April 2022. SARS-CoV-2 spike protein IgG antibody titers were measured using chemiluminescence immunoassay. Data on demographic, clinical, and laboratory characteristics were collected, and patients were characterized by the history of Coronavirus disease 2019 (COVID-19) infection in the past and the number of vaccine doses received. Predictors of antibody response were obtained using multivariate regression analysis. RESULTS: S1/S2 IgG anti-SARS-CoV-2 antibodies were detected in 197 (87.94%) of 224 KTRs with a median [IQR] titers of 307.5 AU/ml [91 AU/ml - 400 AU/ml]. Neutralizing range antibody titers were found in 170/224 (75.9%) KTRs. Diabetes at the time of vaccination was associated with poorer antibody response (aOR 0.31, 95% confidence interval [CI] - 0.10, 0.90; p = 0.032) and vaccination with Covishield™ (ChAdOx1 nCoV- 19 Recombinant CoronaVirus Vaccine) showed higher antibody response as compared to Covaxin™ (BBV152) (aOR 5.04, 95% CI - 1.56, 16.22; p = 0.007). Graft dysfunction at baseline was associated with poorer antibody response. CONCLUSIONS: KTRs showed good antibody response after SARS-CoV-2 vaccination with non-mRNA vaccines. Diabetes and graft dysfunction were associated with poor seroconversion rates.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Kidney Transplantation , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Transplant Recipients , Humans , Kidney Transplantation/adverse effects , Male , Female , Middle Aged , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Adult , Immunoglobulin G/blood , Antibody Formation/immunology , Aged , Vaccination
13.
Clin Immunol ; 266: 110333, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089348

ABSTRACT

Understanding the molecular mechanisms underpinning diverse vaccination responses is critical for developing efficient vaccines. Molecular subtyping can offer insights into heterogeneous nature of responses and aid in vaccine design. We analyzed multi-omic data from 62 haemagglutinin seasonal influenza vaccine recipients (2019-2020), including transcriptomics, proteomics, glycomics, and metabolomics data collected pre-vaccination. We performed a subtyping analysis on the integrated data revealing five subtypes with distinct molecular signatures. These subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining the effectiveness of seasonal vaccines. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.


Subject(s)
Antibodies, Viral , Influenza Vaccines , Influenza, Human , Multiomics , Vaccination , Humans , Adaptive Immunity/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibody Formation/immunology , Hemagglutination Inhibition Tests , Immunity, Innate/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Proteomics/methods , Seasons
14.
Article in English | MEDLINE | ID: mdl-39146978

ABSTRACT

Vaccines targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been pivotal in curtailing the spread of infection. Health care workers, as frontline responders, were among the first to receive vaccination to mitigate coronavirus disease in 2019 (COVID-19) transmission. This study aimed to assess the humoral response elicited by mRNA vaccines, specifically measuring antibodies against the spike S1 protein, a marker of immune response. A cohort of 649 health care workers received three doses of mRNA vaccine, with antibody levels evaluated before and after each dose within a 2- to 3-week interval. Participants were stratified into groups based on prior exposure to the virus: those without prior contact (440 individuals) and those with a history of infection (209 individuals). Among the latter, cases of SARS-CoV-2 infection ranged from asymptomatic (92 individuals) to mild symptomatic (117 individuals). Participants with a history of infection exhibited elevated levels of IgG antibodies against the S1 protein prior to vaccination. Notably, both immunoglobulin IgA class (IgA) and immunoglobulin IgG class (IgG) antibody responses increased significantly post-vaccination, peaking after the second dose for IgG and after the third dose for IgA. Interestingly, the immune response to the vaccine did not vary significantly based on the symptomatic or asymptomatic nature of prior infection. Furthermore, the study findings indicate that completion of the vaccination regimen led to sustained antibody production lasting between 6 months and 9 months. This study underscores the robust and enduring humoral response elicited by mRNA vaccines, particularly among health care workers, irrespective of prior SARS-CoV-2 exposure.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Health Personnel , Immunity, Humoral , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccination , Adult , Female , Humans , Male , Middle Aged , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Formation/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , mRNA Vaccines , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
15.
J Virol ; 98(9): e0076624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194245

ABSTRACT

Antibody responses to influenza vaccines tend to be focused on epitopes encountered during prior influenza exposures, with little production of de novo responses to novel epitopes. To examine the contribution of circulating antibodies to this phenomenon, we passively transferred a hemagglutinin (HA)-specific monoclonal antibody (mAb) into mice before immunizing with whole inactivated virions. The HA mAb inhibited de novo HA-specific antibodies, plasmablasts, germinal center B cells, and memory B cells, while responses to a second antigen in the vaccine, neuraminidase (NA), were uninhibited. The HA mAb potently inhibited de novo antibody responses against epitopes near the HA mAb binding site. The HA mAb also promoted IgG1 class switching, an effect that, unlike the inhibition of HA responses, relied on signaling through Fc-gamma receptors. These studies suggest that circulating antibodies inhibit de novo B cell responses in an antigen-specific manner, which likely contributes to differences in antibody specificities elicited during primary and secondary influenza virus exposures.IMPORTANCEMost humans are exposed to influenza viruses in childhood and then subsequently exposed to antigenically drifted influenza variants later in life. It is unclear if antibodies elicited by earlier influenza virus exposures impact immunity against new influenza virus strains. Here, we used a mouse model to investigate how an anti-hemagglutinin (HA) monoclonal antibody (mAb) affects de novo B cell and antibody responses to the protein targeted by the monoclonal antibody (HA) and a second protein not targeted by the monoclonal antibody [neuraminidase (NA)]. Collectively, our studies suggest that circulating anti-influenza virus antibodies can potently modulate the magnitude and specificity of antibody responses elicited by secondary influenza virus exposures.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , B-Lymphocytes , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Animals , Mice , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , B-Lymphocytes/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Female , Neuraminidase/immunology , Mice, Inbred BALB C , Immunoglobulin G/immunology , Epitopes/immunology , Antibody Formation/immunology
16.
J Proteome Res ; 23(9): 3944-3957, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39146476

ABSTRACT

Solid organ transplant recipients with immunosuppressant regimens to prevent rejection are less able to mount effective immune responses to pathogenic infection. Here, we apply a recently reported mass spectrometry-based serological approach known as Ig-MS to characterize immune responses against infection with SARS-CoV-2 in cohorts of transplant recipients and immunocompetent controls, both at a single early time point following COVID-19 diagnosis as well as over the course of one-month postdiagnosis. We found that the antibody repertoires generated by transplant recipients against SARS-CoV-2 do not differ significantly compared to immunocompetent individuals with regard to repertoire titer, clonality, or glycan composition. Importantly, our study is the first to characterize the evolution of antibody glycan profiles in transplant recipients with COVID-19 disease, presenting evidence that the evolution of glycan composition in these immunocompromised individuals is similar to that in immunocompetent people.


Subject(s)
Antibodies, Viral , COVID-19 , Mass Spectrometry , SARS-CoV-2 , Transplant Recipients , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/diagnosis , SARS-CoV-2/immunology , Antibodies, Viral/blood , Mass Spectrometry/methods , Immunocompromised Host , Middle Aged , Male , Female , Polysaccharides/immunology , Antibody Formation , Adult , Aged
17.
Viral Immunol ; 37(7): 337-345, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39149804

ABSTRACT

Global investment in developing COVID-19 vaccines has been substantial, but vaccine hesitancy has emerged due to misinformation. Concerns about adverse events, vaccine shortages, dosing confusion, mixing vaccines, and access issues contribute to hesitancy. Initially, the WHO recommended homologous vaccination (same vaccine for both doses), but evolving factors led to consideration of heterologous vaccination (different vaccines). The study compared reactogenicity and antibody response for both viral protein spike (S) and nucleocapsid (N) in 205 participants who received three vaccination regimens: same vaccine for all doses (Pfizer), two initial doses of the same vaccine (CoronaVac or AstraZeneca), and a Pfizer booster. ChAdOx1 and BNT162b2 vaccines were the most reactogenic vaccines, while CoronaVac vaccine was the least. ChAdOx1 and BNT162b2 achieved 100% of S-IgG seropositivity with one dose, while CoronaVac required two doses, emphasizing the importance of the second dose in achieving complete immunization across the population with different vaccine regimes. Pfizer recipients showed the highest S-IgG antibody titers, followed by AstraZeneca recipients, both after the first and second doses. A third vaccine dose was essential to boost the S-IgG antibodies and equalize the antibody levels among the different vaccine schedules. CoronaVac induced N-IgG antibodies, while in the Pfizer and AstraZeneca groups, they were induced by a natural infection, reinforcing the role of N protein as a biomarker of infection.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization Schedule , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Antibodies, Viral/blood , Antibody Formation/immunology , BNT162 Vaccine/administration & dosage , BNT162 Vaccine/immunology , ChAdOx1 nCoV-19/immunology , ChAdOx1 nCoV-19/administration & dosage , Coronavirus Nucleocapsid Proteins/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/administration & dosage , Immunization, Secondary , Immunogenicity, Vaccine , Immunoglobulin G/blood , Spike Glycoprotein, Coronavirus/immunology , Vaccination/adverse effects
18.
Nat Commun ; 15(1): 6712, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112489

ABSTRACT

Development of a vaccine against gonorrhoea is a global priority, driven by the rise in antibiotic resistance. Although Neisseria gonorrhoeae (Ng) infection does not induce substantial protective immunity, highly exposed individuals may develop immunity against re-infection with the same strain. Retrospective epidemiological studies have shown that vaccines containing Neisseria meningitidis (Nm) outer membrane vesicles (OMVs) provide a degree of cross-protection against Ng infection. We conducted a clinical trial (NCT04297436) of 4CMenB (Bexsero, GSK), a licensed Nm vaccine containing OMVs and recombinant antigens, comprising a single arm, open label study of two doses with 50 adults in coastal Kenya who have high exposure to Ng. Data from a Ng antigen microarray established that serum IgG and IgA reactivities against the gonococcal homologs of the recombinant antigens in the vaccine peaked at 10 but had declined by 24 weeks. For most reactive OMV-derived antigens, the reverse was the case. A cohort of similar individuals with laboratory-confirmed gonococcal infection were compared before, during, and after infection: their reactivities were weaker and differed from the vaccinated cohort. We conclude that the cross-protection of the 4CMenB vaccine against gonorrhoea could be explained by cross-reaction against a diverse selection of antigens derived from the OMV component.


Subject(s)
Antibodies, Bacterial , Gonorrhea , Immunoglobulin A , Immunoglobulin G , Neisseria gonorrhoeae , Vaccination , Humans , Gonorrhea/immunology , Gonorrhea/prevention & control , Neisseria gonorrhoeae/immunology , Adult , Immunoglobulin A/immunology , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Female , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Kenya/epidemiology , Meningococcal Vaccines/immunology , Meningococcal Vaccines/administration & dosage , Young Adult , Antigens, Bacterial/immunology , Neisseria meningitidis/immunology , Antibody Formation/immunology , Cross Protection/immunology , Middle Aged
19.
Skin Res Technol ; 30(8): e13875, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39120064

ABSTRACT

BACKGROUND: Recent studies increasingly suggest that microbial infections and the immune responses they elicit play significant roles in the pathogenesis of chronic inflammatory skin diseases. This study uses Mendelian randomization (MR) and Bayesian weighted Mendelian randomization (BWMR) to explore the causal relationships between immune antibody responses and four common skin diseases: psoriasis, atopic dermatitis (AD), rosacea, and vitiligo. METHODS: We utilized summary statistics from genome-wide association studies (GWAS) for antibody responses to 13 infectious pathogens and four skin diseases. Single nucleotide polymorphisms (SNPs) were selected as instrumental variables (IVs) to assess causal relationships using multiple MR methods, including inverse variance weighted (IVW), MR Egger, and weighted median. BWMR was also employed to confirm findings and address potential pleiotropy. RESULTS: The IVW analysis identified significant associations between specific antibody responses and the skin diseases studied. Key findings include protective associations of anti-Epstein-Barr virus (EBV) IgG seropositivity and Helicobacter pylori UREA antibody levels with psoriasis and AD. anti-chlamydia trachomatis IgG seropositivity, anti-polyomavirus 2 IgG seropositivity, and varicella zoster virus glycoprotein E and I antibody levels were negatively associated with rosacea, while EBV Elevated levels of the early antigen (EA-D) antibody levels and HHV-6 IE1B antibody levels were positively associated with rosacea. H. pylori Catalase antibody levels were protectively associated with vitiligo, whereas anti-herpes simplex virus 2 (HSV-2) IgG seropositivity was positively associated with vitiligo. The BWMR analysis confirmed these associations. CONCLUSION: This study underscores the significant role of H. pylori and other pathogens in these skin diseases, suggesting both protective and exacerbating effects depending on the specific condition. Understanding these pathogen-immune interactions can lead to the development of more effective, personalized treatments and preventative strategies, ultimately improving patient outcomes and quality of life.


Subject(s)
Bayes Theorem , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Dermatitis, Atopic/immunology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/blood , Rosacea/immunology , Rosacea/genetics , Vitiligo/genetics , Vitiligo/immunology , Antibody Formation/genetics , Psoriasis/immunology , Psoriasis/genetics , Skin Diseases/immunology , Skin Diseases/genetics
20.
Diagn Microbiol Infect Dis ; 110(2): 116425, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098282

ABSTRACT

The study aimed to evaluate the effect of booster dose COVID-19 vaccines on prevention and humoral immune response in individuals with different vaccination schemes during the period BA.4 and BA.5 omicron sub-variants were globally dominant. The study included 146 individuals who preferred different vaccination schemes for booster doses. Anti-spike/RBD-IgG and neutralizing antibody levels were measured 28 days after the booster dose vaccination upon their consent. There is no significant difference between median antibody titers detected according to different vaccination schemes. SARS-CoV-2 neutralizing antibody inhibition percentages were detected significantly higher in serum samples before and after the last booster dose in 2 BNT162b2+1 BNT162b2(99.42 %), 2 BNT162b2 + 2 BNT162b2(99.42 %), and 2 BNT162b2 + 3 BNT162b2(99.42 %) vaccination schemes (p = 0.004, p = 0.044, p = 0.002,respectively). The study indicated that a booster vaccination dose provides a high level of protection against severe COVID-19 and death. We think that the variant-specific pancoronavirus vaccines will be necessary to protect against breakthrough infections.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Humans , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/blood , Male , Female , SARS-CoV-2/immunology , Prospective Studies , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Middle Aged , Antibodies, Neutralizing/blood , Adult , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Aged , Immunoglobulin G/blood , Vaccination , Spike Glycoprotein, Coronavirus/immunology , Antibody Formation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL