Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.373
Filter
1.
Microb Biotechnol ; 17(7): e14518, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38953907

ABSTRACT

Porcine epidemic diarrhoea virus (PEDV) infects pigs of all ages by invading small intestine, causing acute diarrhoea, vomiting, and dehydration with high morbidity and mortality among newborn piglets. However, current PEDV vaccines are not effective to protect the pigs from field epidemic strains because of poor mucosal immune response and strain variation. Therefore, it is indispensable to develop a novel oral vaccine based on epidemic strains. Bacillus subtilis spores are attractive delivery vehicles for oral vaccination on account of the safety, high stability, and low cost. In this study, a chimeric gene CotC-Linker-COE (CLE), comprising of the B. subtilis spore coat gene cotC fused to the core neutralizing epitope CO-26 K equivalent (COE) of the epidemic strain PEDV-AJ1102 spike protein gene, was constructed. Then recombinant B. subtilis displaying the CLE on the spore surface was developed by homologous recombination. Mice were immunized by oral route with B. subtilis 168-CLE, B. subtilis 168, or phosphate-buffered saline (PBS) as control. Results showed that the IgG antibodies and cytokine (IL-4, IFN-γ) levels in the B. subtilis 168-CLE group were significantly higher than the control groups. This study demonstrates that B. subtilis 168-CLE can generate specific systemic immune and mucosal immune responses and is a potential vaccine candidate against PEDV infection.


Subject(s)
Antibodies, Viral , Bacillus subtilis , Porcine epidemic diarrhea virus , Spores, Bacterial , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/immunology , Animals , Bacillus subtilis/genetics , Bacillus subtilis/immunology , Spores, Bacterial/genetics , Spores, Bacterial/immunology , Mice , Antibodies, Viral/blood , Swine , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Vaccines/administration & dosage , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/microbiology , Swine Diseases/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Administration, Oral , Cytokines/metabolism , Immunoglobulin G/blood , Mice, Inbred BALB C , Female , Cell Surface Display Techniques , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
2.
Commun Biol ; 7(1): 805, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961245

ABSTRACT

Precise epitope determination of therapeutic antibodies is of great value as it allows for further comprehension of mechanism of action, therapeutic responsiveness prediction, avoidance of unwanted cross reactivity, and vaccine design. The golden standard for discontinuous epitope determination is the laborious X-ray crystallography method. Here, we present a combinatorial method for rapid mapping of discontinuous epitopes by mammalian antigen display, eliminating the need for protein expression and purification. The method is facilitated by automated workflows and tailored software for antigen analysis and oligonucleotide design. These oligos are used in automated mutagenesis to generate an antigen receptor library displayed on mammalian cells for direct binding analysis by flow cytometry. Through automated analysis of 33930 primers an optimized single condition cloning reaction was defined allowing for mutation of all surface-exposed residues of the receptor binding domain of SARS-CoV-2. All variants were functionally expressed, and two reference binders validated the method. Furthermore, epitopes of three novel therapeutic antibodies were successfully determined followed by evaluation of binding also towards SARS-CoV-2 Omicron BA.2. We find the method to be highly relevant for rapid construction of antigen libraries and determination of antibody epitopes, especially for the development of therapeutic interventions against novel pathogens.


Subject(s)
COVID-19 , Epitope Mapping , Epitopes , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Epitope Mapping/methods , Epitopes/immunology , Epitopes/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/immunology , COVID-19/virology , Peptide Library , Antibodies, Viral/immunology , Animals , HEK293 Cells , Cell Surface Display Techniques/methods , Gene Library
3.
J Med Virol ; 96(7): e29778, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38965882

ABSTRACT

Active and passive immunization is used in high-risk patients to prevent severe courses of COVID-19, but the impact of prophylactic neutralizing antibodies on the immune reaction to the mRNA vaccines has remained enigmatic. Here we show that CD4 T and B cell responses to Spikevax booster immunization are suppressed by the therapeutic antibodies Casirivimab and Imdevimab. B cell and T cell responses were significantly induced in controls but not in antibody-treated patients. The data indicates that humoral immunity, i. e. high levels of antibodies, negatively impacts reactive immunity, resulting in blunted cellular responses upon boosting. This argues for temporal separation of vaccination efforts; with active vaccination preferably applied before prophylactic therapeutic antibody treatment.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , B-Lymphocytes , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/prevention & control , COVID-19/immunology , B-Lymphocytes/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , Middle Aged , Male , Female , Vaccination , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , Immunization, Secondary , Immunity, Humoral , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use
4.
Nat Commun ; 15(1): 5589, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961063

ABSTRACT

As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1-/- mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate.


Subject(s)
Administration, Intranasal , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Female , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Mice , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Cricetinae , Humans , Measles-Mumps-Rubella Vaccine/immunology , Measles-Mumps-Rubella Vaccine/administration & dosage , Measles virus/immunology , Measles virus/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mumps virus/immunology , Mumps virus/genetics , Mice, Knockout , Mesocricetus , Immunoglobulin A/immunology , Immunoglobulin A/blood
5.
Front Immunol ; 15: 1406138, 2024.
Article in English | MEDLINE | ID: mdl-38975334

ABSTRACT

Heterologous prime-boost has broken the protective immune response bottleneck of the COVID-19 vaccines. however, the underlying mechanisms have not been fully elucidated. Here, we investigated antibody responses and explored the response of germinal center (GC) to priming with inactivated vaccines and boosting with heterologous adenoviral-vectored vaccines or homologous inactivated vaccines in mice. Antibody responses were dramatically enhanced by both boosting regimens. Heterologous immunization induced more robust GC activation, characterized by increased Tfh cell populations and enhanced helper function. Additionally, increased B-cell activation and antibody production were observed in a heterologous regimen. Libra-seq was used to compare the differences of S1-, S2- and NTD-specific B cells between homologous and heterologous vaccination, respectively. S2-specific CD19+ B cells presented increased somatic hypermutations (SHMs), which were mainly enriched in plasma cells. Moreover, a heterologous booster dose promoted the clonal expansion of B cells specific to S2 and NTD regions. In conclusion, the functional role of Tfh and B cells following SARS-CoV-2 heterologous vaccination may be important for modulating antibody responses. These findings provide new insights for the development of SARS-CoV-2 vaccines that induce more robust antibody response.


Subject(s)
Antibodies, Viral , Antibody Formation , B-Lymphocytes , COVID-19 Vaccines , COVID-19 , Germinal Center , Immunization, Secondary , SARS-CoV-2 , T Follicular Helper Cells , Animals , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , B-Lymphocytes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Mice , COVID-19/immunology , COVID-19/prevention & control , T Follicular Helper Cells/immunology , Germinal Center/immunology , Antibody Formation/immunology , Female , Somatic Hypermutation, Immunoglobulin , Vaccination , Mice, Inbred BALB C , Humans , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics
6.
Methods Mol Biol ; 2829: 185-194, 2024.
Article in English | MEDLINE | ID: mdl-38951334

ABSTRACT

Insect cell expression has been successfully used for the production of viral antigens as part of commercial vaccine development. As expression host, insect cells offer advantage over bacterial system by presenting the ability of performing post-translational modifications (PTMs) such as glycosylation and phosphorylation thus preserving the native functionality of the proteins especially for viral antigens. Insect cells have limitation in exactly mimicking some proteins which require complex glycosylation pattern. The recent advancement in insect cell engineering strategies could overcome this limitation to some extent. Moreover, cost efficiency, timelines, safety, and process adoptability make insect cells a preferred platform for production of subunit antigens for human and animal vaccines. In this chapter, we describe the method for producing the SARS-CoV2 spike ectodomain subunit antigen for human vaccine development and the virus like particle (VLP), based on capsid protein of porcine circovirus virus 2 (PCV2d) antigen for animal vaccine development using two different insect cell lines, SF9 & Hi5, respectively. This methodology demonstrates the flexibility and broad applicability of insect cell as expression host.


Subject(s)
Antigens, Viral , Baculoviridae , Spike Glycoprotein, Coronavirus , Animals , Baculoviridae/genetics , Antigens, Viral/genetics , Antigens, Viral/immunology , Sf9 Cells , Humans , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Recombinant Proteins/genetics , Cell Line , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/biosynthesis , Capsid Proteins/genetics , Capsid Proteins/immunology , Glycosylation , Insecta/genetics , Spodoptera , COVID-19 Vaccines/genetics , COVID-19 Vaccines/immunology
7.
Cell Death Dis ; 15(6): 458, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38937437

ABSTRACT

SARS-CoV-2 infection is initiated by Spike glycoprotein binding to the human angiotensin-converting enzyme 2 (ACE2) receptor via its receptor binding domain. Blocking this interaction has been proven to be an effective approach to inhibit virus infection. Here we report the discovery of a neutralizing nanobody named VHH60, which was directly produced from an engineering nanobody library based on a commercialized nanobody within a very short period. VHH60 competes with human ACE2 to bind the receptor binding domain of the Spike protein at S351, S470-471and S493-494 as determined by structural analysis, with an affinity of 2.56 nM. It inhibits infections of both ancestral SARS-CoV-2 strain and pseudotyped viruses harboring SARS-CoV-2 wildtype, key mutations or variants at the nanomolar level. Furthermore, VHH60 suppressed SARS-CoV-2 infection and propagation 50-fold better and protected mice from death for twice as long as the control group after SARS-CoV-2 nasal infections in vivo. Therefore, VHH60 is not only a powerful nanobody with a promising profile for disease control but also provides evidence for a highly effective and rapid approach to generating therapeutic nanobodies.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , COVID-19 , SARS-CoV-2 , Single-Domain Antibodies , Spike Glycoprotein, Coronavirus , SARS-CoV-2/immunology , SARS-CoV-2/drug effects , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/immunology , Humans , Animals , COVID-19/immunology , COVID-19/virology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Mice , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Neutralizing/pharmacology , COVID-19 Drug Treatment , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , HEK293 Cells , Mice, Inbred BALB C , Protein Binding , Female
8.
PLoS One ; 19(6): e0303303, 2024.
Article in English | MEDLINE | ID: mdl-38900738

ABSTRACT

BACKGROUND: Understanding the relative contributions of SARS-CoV-2 infection-induced and vaccine-induced seroprevalence is key to measuring overall population-level seroprevalence and help guide policy decisions. METHODS: Using a series of six population-based cross-sectional surveys conducted among persons aged ≥7 years in a large health system with over 4.5 million members between May 2021 and April 2022, we combined data from the electronic health record (EHR), an electronic survey and SARS-CoV-2 spike antibody binding assay, to assess the relative contributions of infection and vaccination to population-level SARS-CoV-2 seroprevalence. EHR and survey data were incorporated to determine spike antibody positivity due to SARS-CoV-2 infection and COVID-19 vaccination. We used sampling and non-response weighting to create population-level estimates. RESULTS: We enrolled 4,319 persons over six recruitment waves. SARS-CoV-2 spike antibody seroprevalence increased from 83.3% (CI 77.0-88.9) in May 2021 to 93.5% (CI 89.5-97.5) in April 2022. By April 2022, 68.5% (CI 61.9-74.3) of the population was seropositive from COVID-19 vaccination only, 13.9% (10.7-17.9) from COVID-19 vaccination and prior diagnosed SARS-CoV-2 infection, 8.2% (CI 4.5-14.5) from prior diagnosed SARS-CoV-2 infection only and 2.9% (CI 1.1-7.6) from prior undiagnosed SARS-CoV-2 infection only. We found high agreement (≥97%) between EHR and survey data for ascertaining COVID-19 vaccination and SARS-CoV-2 infection status. CONCLUSIONS: By April 2022, 93.5% of persons had detectable SARS-CoV-2 spike antibody, predominantly from COVID-19 vaccination. In this highly vaccinated population and over 18 months into the pandemic, SARS-CoV-2 infection without COVID-19 vaccination was a small contributor to overall population-level seroprevalence.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/epidemiology , COVID-19/immunology , COVID-19/prevention & control , Seroepidemiologic Studies , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , Male , Antibodies, Viral/blood , Antibodies, Viral/immunology , Middle Aged , Spike Glycoprotein, Coronavirus/immunology , Female , Adult , Aged , Cross-Sectional Studies , Adolescent , Child , Young Adult , Vaccination , Aged, 80 and over
9.
Sci Immunol ; 9(96): eadj8526, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905326

ABSTRACT

Memory CD4 T cells are critical to human immunity, yet it is unclear whether viral inflammation during memory formation has long-term consequences. Here, we compared transcriptional and epigenetic landscapes of Spike (S)-specific memory CD4 T cells in 24 individuals whose first exposure to S was via SARS-CoV-2 infection or mRNA vaccination. Nearly 2 years after memory formation, S-specific CD4 T cells established by infection remained enriched for transcripts related to cytotoxicity and for interferon-stimulated genes, likely because of a chromatin accessibility landscape altered by inflammation. Moreover, S-specific CD4 T cells primed by infection had reduced proliferative capacity in vitro relative to vaccine-primed cells. Furthermore, the transcriptional state of S-specific memory CD4 T cells was minimally altered by booster immunization and/or breakthrough infection. Thus, infection-associated inflammation durably imprints CD4 T cell memory, which affects the function of these cells and may have consequences for long-term immunity.


Subject(s)
CD4-Positive T-Lymphocytes , COVID-19 , Immunologic Memory , Inflammation , Memory T Cells , SARS-CoV-2 , Humans , COVID-19/immunology , SARS-CoV-2/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Inflammation/immunology , Memory T Cells/immunology , Spike Glycoprotein, Coronavirus/immunology , Female , Male , Adult , COVID-19 Vaccines/immunology
10.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38928018

ABSTRACT

An estimation of the proportion of nonsynonymous to synonymous mutation (dn/ds, ω) of the SARS-CoV-2 genome would indicate the evolutionary dynamics necessary to evolve into novel strains with increased infection, virulence, and vaccine neutralization. A temporal estimation of ω of the whole genome, and all twenty-nine SARS-CoV-2 genes of major virulent strains of alpha, delta and omicron demonstrates that the SARS-CoV-2 genome originally emerged (ω ~ 0.04) with a strong purifying selection (ω < 1) and reached (ω ~ 0.85) in omicron towards diversifying selection (ω > 1). A marked increase in the ω occurred in the spike gene from alpha (ω = 0.2) to omicron (ω = 1.97). The ω of the replication machinery genes including RDRP, NSP3, NSP4, NSP7, NSP8, NSP10, NSP13, NSP14, and ORF9 are markedly increased, indicating that these genes/proteins are yet to be evolutionary stabilized and are contributing to the evolution of novel virulent strains. The delta-specific maximum increase in ω in the immunomodulatory genes of NSP8, NSP10, NSP16, ORF4, ORF5, ORF6, ORF7A, and ORF8 compared to alpha or omicron indicates delta-specific vulnerabilities for severe COVID-19 related hospitalization and death. The maximum values of ω are observed for spike (S), NSP4, ORF8 and NSP15, which indicates that the gene-specific temporal estimation of ω identifies specific genes for its super-infectivity and virulency that could be targeted for drug development.


Subject(s)
COVID-19 Vaccines , COVID-19 , Evolution, Molecular , Genome, Viral , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/immunology , Humans , COVID-19 Vaccines/immunology , COVID-19/virology , COVID-19/immunology , Virulence/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Mutation , Phylogeny
11.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928216

ABSTRACT

The pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a global health concern. Cell entry of SARS-CoV-2 depends on viral spike (S) proteins binding to cellular receptors (ACE2) and their subsequent priming by host cell proteases (TMPRSS2). Assessing effects of viral-induced host response factors and determining which cells are used by SARS-CoV-2 for entry might provide insights into viral transmission, add clarity to the virus' pathogenesis, and possibly reveal therapeutic targets. Mast cells (MCs) are ubiquitously expressed tissue cells that act as immune sentinels given their ability to react specifically to pathogens at environmental interfaces, such as in the lung. Several lines of evidence suggest a critical role for MCs in SARS-CoV-2 infections based on patients' mediator profiles, especially the "cytokine storm" responsible for most morbidity and mortality. In this pilot study, we demonstrated that human lung MCs (n = 3 donors) are a source of renin and that they upregulate the membrane receptor for SARS-CoV-2 (ACE2) as well as the protease required for cellular entry (TMPRSS2) under certain conditions. We hypothesized that infection of human MCs with SARS-CoV-2 may be a heretofore-unrecognized mechanism of viral pathogenesis, and further studies are required to assess this question.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Lung , Mast Cells , SARS-CoV-2 , Serine Endopeptidases , Humans , Mast Cells/virology , Mast Cells/immunology , Mast Cells/metabolism , SARS-CoV-2/pathogenicity , SARS-CoV-2/immunology , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/immunology , COVID-19/pathology , Angiotensin-Converting Enzyme 2/metabolism , Lung/virology , Lung/pathology , Lung/immunology , Serine Endopeptidases/metabolism , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology
12.
J Clin Immunol ; 44(6): 147, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38856804

ABSTRACT

PURPOSE: Asymptomatic SARS-CoV-2 infections were widely reported during the COVID-19 pandemic, acting as a hidden source of infection. Many existing studies investigating asymptomatic immunity failed to recruit true asymptomatic individuals. Thus, we conducted a longitudinal cohort study to evaluate humoral- and cell-mediated responses to infection and vaccination in well-defined asymptomatic young adults (the Asymptomatic COVID-19 in Education [ACE] cohort). METHODS: Asymptomatic testing services located at three UK universities identified asymptomatic young adults who were subsequently recruited with age- and sex-matched symptomatic and uninfected controls. Blood and saliva samples were collected after SARS-CoV-2 Wuhan infection, and again after vaccination. 51 participant's anti-spike antibody titres, neutralizing antibodies, and spike-specific T-cell responses were measured, against both Wuhan and Omicron B.1.1.529.1. RESULTS: Asymptomatic participants exhibited reduced Wuhan-specific neutralization antibodies pre- and post-vaccination, as well as fewer Omicron-specific neutralization antibodies post-vaccination, compared to symptomatic participants. Lower Wuhan and Omicron-specific IgG titres in asymptomatic individuals were also observed pre- and post-vaccination, compared to symptomatic participants. There were no differences in salivary IgA levels. Conventional flow cytometry analysis and multi-dimensional clustering analysis indicated unvaccinated asymptomatic participants had significantly fewer Wuhan-specific IL-2 secreting CD4+ CD45RA+ T cells and activated CD8+ T cells than symptomatic participants, though these differences dissipated after vaccination. CONCLUSIONS: Asymptomatic infection results in decreased antibody and T cell responses to further exposure to SARS-CoV-2 variants, compared to symptomatic infection. Post-vaccination, antibody responses are still inferior, but T cell immunity increases to match symptomatic subjects, emphasising the importance of vaccination to help protect asymptomatic individuals against future variants.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Asymptomatic Infections , COVID-19 , Immunity, Cellular , Immunity, Humoral , SARS-CoV-2 , Humans , COVID-19/immunology , SARS-CoV-2/immunology , Male , Female , Antibodies, Viral/blood , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Young Adult , Adult , COVID-19 Vaccines/immunology , Cohort Studies , Longitudinal Studies , Vaccination , Immunoglobulin G/blood , Immunoglobulin G/immunology , United Kingdom/epidemiology , Adolescent , Spike Glycoprotein, Coronavirus/immunology
13.
BMC Infect Dis ; 24(1): 584, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867165

ABSTRACT

BACKGROUND: Natural infection and vaccination against SARS-CoV-2 is associated with the development of immunity against the structural proteins of the virus. Specifically, the two most immunogenic are the S (spike) and N (nucleocapsid) proteins. Seroprevalence studies performed in university students provide information to estimate the number of infected patients (symptomatic or asymptomatic) and generate knowledge about the viral spread, vaccine efficacy, and epidemiological control. Which, the aim of this study was to evaluate IgG antibodies against the S and N proteins of SARS-CoV-2 at university students from Southern Mexico. METHODS: A total of 1418 serum samples were collected from eighteen work centers of the Autonomous University of Guerrero. Antibodies were detected by Indirect ELISA using as antigen peptides derived from the S and N proteins. RESULTS: We reported a total seroprevalence of 39.9% anti-S/N (positive to both antigens), 14.1% anti-S and 0.5% anti-N. The highest seroprevalence was reported in the work centers from Costa Grande, Acapulco and Centro. Seroprevalence was associated with age, COVID-19, contact with infected patients, and vaccination. CONCLUSION: University students could play an essential role in disseminating SARS-CoV-2. We reported a seroprevalence of 54.5% against the S and N proteins, which could be due to the high population rate and cultural resistance to safety measures against COVID-19 in the different regions of the state.


Subject(s)
Antibodies, Viral , COVID-19 , Coronavirus Nucleocapsid Proteins , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Students , Humans , Mexico/epidemiology , Male , Female , Cross-Sectional Studies , Spike Glycoprotein, Coronavirus/immunology , Immunoglobulin G/blood , COVID-19/epidemiology , COVID-19/immunology , Young Adult , Antibodies, Viral/blood , SARS-CoV-2/immunology , Seroepidemiologic Studies , Adult , Universities , Coronavirus Nucleocapsid Proteins/immunology , Adolescent , Phosphoproteins/immunology
14.
Front Immunol ; 15: 1397052, 2024.
Article in English | MEDLINE | ID: mdl-38911866

ABSTRACT

Background: Immunocompromised patients are at particular risk of Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) infection and previous findings suggest that the infection or vaccination induced immune response decreases over time. Our main goal was to investigate the SARS-CoV-2-specific immune response in rheumatoid arthritis patients and healthy controls over prolonged time. Methods: The SARS-CoV-2-specific humoral immune response was measured by Elecsys Anti-SARS-CoV-2 Spike (S) immunoassay, and antibodies against SARS-CoV-2 nucleocapsid protein (NCP) were also evaluated by Euroimmun enzyme-linked immunosorbent assay (ELISA) test. The SARS-CoV-2-specific T-cell response was detected by an IFN- γ release assay. Results: We prospectively enrolled 84 patients diagnosed with rheumatoid arthritis (RA) and 43 healthy controls in our longitudinal study. Our findings demonstrate that RA patients had significantly lower anti-S antibody response and reduced SARS-CoV-2-specific T-cell response compared to healthy controls (p<0.01 for healthy controls, p<0.001 for RA patients). Furthermore, our results present evidence of a notable increase in the SARS-CoV-2-specific humoral immune response during the follow-up period in both study groups (p<0.05 for healthy volunteers, p<0.0001 for RA patients, rank-sum test). Participants who were vaccinated against Coronavirus disease-19 (COVID-19) during the interim period had 2.72 (CI 95%: 1.25-5.95, p<0.05) times higher anti-S levels compared to those who were not vaccinated during this period. Additionally, individuals with a confirmed SARS-CoV-2 infection exhibited 2.1 times higher (CI 95%: 1.31-3.37, p<0.01) anti-S levels compared to those who were not infected during the interim period. It is worth noting that patients treated with targeted therapy had 52% (CI 95%: 0.25-0.94, p<0.05) lower anti-S levels compared to matched patients who did not receive targeted therapy. Concerning the SARS-CoV-2-specific T-cell response, our findings revealed that its level had not changed substantially in the study groups. Conclusion: Our present data revealed that the level of SARS-CoV-2-specific humoral immune response is actually higher, and the SARS-CoV-2-specific T-cell response remained at the same level over time in both study groups. This heightened humoral response, the nearly permanent SARS-CoV-2-specific T-cell response and the coexistence of different SARS-CoV-2 variants within the population, might be contributing to the decline in severe COVID-19 cases.


Subject(s)
Antibodies, Viral , Arthritis, Rheumatoid , COVID-19 , Immunity, Humoral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Arthritis, Rheumatoid/immunology , SARS-CoV-2/immunology , Male , Female , Middle Aged , COVID-19/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Aged , Spike Glycoprotein, Coronavirus/immunology , Adult , T-Lymphocytes/immunology , Coronavirus Nucleocapsid Proteins/immunology , Prospective Studies , Phosphoproteins/immunology , Case-Control Studies , Longitudinal Studies
15.
Bull Exp Biol Med ; 176(6): 776-780, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38896316

ABSTRACT

We studied a needle-free jet injection delivery of an experimental mRNA vaccine encoding the receptor-binding domain of the SARS-CoV-2 S protein (mRNA-RBD). Immunization of BALB/c mice with mRNA-RBD by a needle-free jet injector induced high levels of antibodies with virus-neutralizing activity and a virus-specific T-cell response. The immune response was low in the group of mice that received intramuscular injection of mRNA-RBD. The effectiveness of this simple and safe method of mRNA delivering has been demonstrated. Thus, jet injection of mRNA vaccine can be a good alternative to lipid nanoparticles.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Mice, Inbred BALB C , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Mice , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Neutralizing/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Injections, Jet , mRNA Vaccines , RNA, Messenger/genetics , RNA, Messenger/immunology , Injections, Intramuscular , Female , Humans , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage
16.
PLoS Pathog ; 20(6): e1011569, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900807

ABSTRACT

Antibodies perform both neutralizing and non-neutralizing effector functions that protect against certain pathogen-induced diseases. A human antibody directed at the SARS-CoV-2 Spike N-terminal domain (NTD), DH1052, was recently shown to be non-neutralizing, yet it protected mice and cynomolgus macaques from severe disease. The mechanisms of NTD non-neutralizing antibody-mediated protection are unknown. Here we show that Fc effector functions mediate NTD non-neutralizing antibody (non-nAb) protection against SARS-CoV-2 MA10 viral challenge in mice. Though non-nAb prophylactic infusion did not suppress infectious viral titers in the lung as potently as neutralizing antibody (nAb) infusion, disease markers including gross lung discoloration were similar in nAb and non-nAb groups. Fc functional knockout substitutions abolished non-nAb protection and increased viral titers in the nAb group. Fc enhancement increased non-nAb protection relative to WT, supporting a positive association between Fc functionality and degree of protection from SARS-CoV-2 infection. For therapeutic administration of antibodies, non-nAb effector functions contributed to virus suppression and lessening of lung discoloration, but the presence of neutralization was required for optimal protection from disease. This study demonstrates that non-nAbs can utilize Fc-mediated mechanisms to lower viral load and prevent lung damage due to coronavirus infection.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Immunoglobulin Fc Fragments , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , SARS-CoV-2/immunology , Mice , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology , Immunoglobulin Fc Fragments/immunology , Spike Glycoprotein, Coronavirus/immunology , Humans , Female , Protein Domains/immunology , Viral Load , Lung/virology , Lung/immunology , Lung/pathology
17.
Nat Immunol ; 25(7): 1158-1171, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38902519

ABSTRACT

Up to 25% of individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibit postacute cognitive sequelae. Although millions of cases of coronavirus disease 2019 (COVID-19)-mediated memory dysfunction are accumulating worldwide, the underlying mechanisms and how vaccination lowers risk are unknown. Interleukin-1 (IL-1), a key component of innate immune defense against SARS-CoV-2 infection, is elevated in the hippocampi of individuals with COVID-19. Here we show that intranasal infection of C57BL/6J mice with SARS-CoV-2 Beta variant leads to central nervous system infiltration of Ly6Chi monocytes and microglial activation. Accordingly, SARS-CoV-2, but not H1N1 influenza virus, increases levels of brain IL-1ß and induces persistent IL-1R1-mediated loss of hippocampal neurogenesis, which promotes postacute cognitive deficits. Vaccination with a low dose of adenoviral-vectored spike protein prevents hippocampal production of IL-1ß during breakthrough SARS-CoV-2 infection, loss of neurogenesis and subsequent memory deficits. Our study identifies IL-1ß as one potential mechanism driving SARS-CoV-2-induced cognitive impairment in a new mouse model that is prevented by vaccination.


Subject(s)
COVID-19 , Hippocampus , Interleukin-1beta , Memory Disorders , Mice, Inbred C57BL , Neurogenesis , SARS-CoV-2 , Animals , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Mice , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Hippocampus/immunology , Hippocampus/metabolism , Memory Disorders/immunology , Neurogenesis/immunology , Vaccination , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Vaccines/immunology , Male , Humans , Microglia/immunology , Microglia/metabolism , Disease Models, Animal , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-1 Type I/genetics , Monocytes/immunology , Monocytes/metabolism , Female
18.
J Immunol Res ; 2024: 9313267, 2024.
Article in English | MEDLINE | ID: mdl-38939745

ABSTRACT

Vaccination is one of the most effective prophylactic public health interventions for the prevention of infectious diseases such as coronavirus disease (COVID-19). Considering the ongoing need for new COVID-19 vaccines, it is crucial to modify our approach and incorporate more conserved regions of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to effectively address emerging viral variants. The nucleocapsid protein is a structural protein of SARS-CoV-2 that is involved in replication and immune responses. Furthermore, this protein offers significant advantages owing to the minimal accumulation of mutations over time and the inclusion of key T-cell epitopes critical for SARS-CoV-2 immunity. A novel strategy that may be suitable for the new generation of vaccines against COVID-19 is to use a combination of antigens, including the spike and nucleocapsid proteins, to elicit robust humoral and potent cellular immune responses, along with long-lasting immunity. The strategic use of multiple antigens aims to enhance vaccine efficacy and broaden protection against viruses, including their variants. The immune response against the nucleocapsid protein from other coronavirus is long-lasting, and it can persist up to 11 years post-infection. Thus, the incorporation of nucleocapsids (N) into vaccine design adds an important dimension to vaccination efforts and holds promise for bolstering the ability to combat COVID-19 effectively. In this review, we summarize the preclinical studies that evaluated the use of the nucleocapsid protein as antigen. This study discusses the use of nucleocapsid alone and its combination with spike protein or other proteins of SARS-CoV-2.


Subject(s)
COVID-19 Vaccines , COVID-19 , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Humans , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/genetics , Immunogenicity, Vaccine , Animals , Phosphoproteins/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Epitopes, T-Lymphocyte/immunology , Antibodies, Viral/immunology , Nucleocapsid Proteins/immunology
19.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892276

ABSTRACT

Heterologous vaccines, which induce immunity against several related pathogens, can be a very useful and rapid way to deal with new pandemics. In this study, the potential impact of licensed COVID-19 vaccines on cytotoxic and helper cell immune responses against Khosta-2, a novel sarbecovirus that productively infects human cells, was analyzed for the 567 and 41 most common HLA class I and II alleles, respectively. Computational predictions indicated that most of these 608 alleles, covering more than 90% of the human population, contain sufficient fully conserved T-cell epitopes between the Khosta-2 and SARS-CoV-2 spike-in proteins. Ninety percent of these fully conserved peptides for class I and 93% for class II HLA molecules were verified as epitopes recognized by CD8+ or CD4+ T lymphocytes, respectively. These results show a very high correlation between bioinformatic prediction and experimental assays, which strongly validates this study. This immunoinformatics analysis allowed a broader assessment of the alleles that recognize these peptides, a global approach at the population level that is not possible with experimental assays. In summary, these findings suggest that both cytotoxic and helper cell immune protection elicited by currently licensed COVID-19 vaccines should be effective against Khosta-2 virus infection. Finally, by being rapidly adaptable to future coronavirus pandemics, this study has potential public health implications.


Subject(s)
COVID-19 Vaccines , COVID-19 , Epitopes, T-Lymphocyte , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/chemistry , Epitopes, T-Lymphocyte/immunology , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Cross Protection/immunology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , HLA Antigens/immunology , HLA Antigens/genetics , Animals
20.
Nat Commun ; 15(1): 5496, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38944664

ABSTRACT

Evolution of SARS-CoV-2 alters the antigenicity of the immunodominant spike (S) receptor-binding domain and N-terminal domain, undermining the efficacy of vaccines and antibody therapies. To overcome this challenge, we set out to develop a vaccine focusing antibody responses on the highly conserved but metastable S2 subunit, which folds as a spring-loaded fusion machinery. We describe a strategy for prefusion-stabilization and high yield recombinant production of SARS-CoV-2 S2 trimers with native structure and antigenicity. We demonstrate that our design strategy is broadly generalizable to sarbecoviruses, as exemplified with the SARS-CoV-1 (clade 1a) and PRD-0038 (clade 3) S2 subunits. Immunization of mice with a prefusion-stabilized SARS-CoV-2 S2 trimer elicits broadly reactive sarbecovirus antibodies and neutralizing antibody titers of comparable magnitude against Wuhan-Hu-1 and the immune evasive XBB.1.5 variant. Vaccinated mice were protected from weight loss and disease upon challenge with XBB.1.5, providing proof-of-principle for fusion machinery sarbecovirus vaccines.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Mice , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , SARS-CoV-2/immunology , Humans , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Female , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...