Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 18.785
Filtrer
1.
Appl Microbiol Biotechnol ; 108(1): 431, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093478

RÉSUMÉ

African swine fever (ASF) is a highly contagious and fatal viral disease that has caused huge economic losses to the pig and related industries worldwide. At present, rapid, accurate, and sensitive laboratory detection technologies are important means of preventing and controlling ASF. However, because attenuated strains of African swine fever virus (ASFV) are constantly emerging, an ASFV antibody could be used more effectively to investigate the virus and control the disease on pig farms. The isolation of ASFV-specific antibodies is also essential for the diagnosis of ASF. Therefore, in this study, we developed two chemiluminescence immunoassays (CLIAs) to detect antibodies directed against ASFV p72: a traditional plate-type blocking CLIA (p72-CLIA) and an automatic tubular competitive CLIA based on magnetic particles (p72-MPCLIA). We compared the diagnostic performance of these two methods to provide a feasible new method for the effective prevention and control of ASF and the purification of ASFV. The cut-off value, diagnostic sensitivity (Dsn), and diagnostic specificity (Dsp) of p72-CLIA were 40%, 100%, and 99.6%, respectively, in known background serum, whereas those of p72-MPCLIA were 36%, 100%, and 99.6%, respectively. Thus, both methods show good Dsn, Dsp, and repeatability. However, when analytical sensitivity was evaluated, p72-MPCLIA was more sensitive than p72-CLIA or a commercial enzyme-linked immunosorbent assay. More importantly, p72-MPCLIA reduced the detection time to 15 min and allowed fully automated detection. In summary, p72-MPCLIA showed superior diagnostic performance and offered a new tool for detecting ASFV infections in the future. KEY POINTS: • Two chemiluminescence immunoassay (plate-type CLIA and tubular CLIA) methods based on p72 monoclonal antibody (mAb) were developed to detect ASFV antibody. • Both methods show good diagnostic performance (Dsn (100%), Dsp (99.6%), and good repeatability), and p72-MPCLIA detects antibodies against ASFV p72 with high efficiency in just 15 min.


Sujet(s)
Virus de la peste porcine africaine , Peste porcine africaine , Anticorps antiviraux , Mesures de luminescence , Sensibilité et spécificité , Virus de la peste porcine africaine/immunologie , Virus de la peste porcine africaine/isolement et purification , Animaux , Peste porcine africaine/diagnostic , Peste porcine africaine/virologie , Peste porcine africaine/immunologie , Suidae , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Dosage immunologique/méthodes , Mesures de luminescence/méthodes
2.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39096433

RÉSUMÉ

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Sujet(s)
Baculoviridae , Capripoxvirus , Test ELISA , Maladies des chèvres , Capra , Protéines de l'enveloppe virale , Capripoxvirus/génétique , Capripoxvirus/isolement et purification , Baculoviridae/génétique , Animaux , Maladies des chèvres/virologie , Maladies des chèvres/diagnostic , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Capra/virologie , Test ELISA/méthodes , Infections à Poxviridae/diagnostic , Infections à Poxviridae/médecine vétérinaire , Infections à Poxviridae/virologie , Protéines recombinantes/génétique , Protéines recombinantes/isolement et purification , Protéines recombinantes/immunologie , Virion/génétique , Protéines du core viral/génétique , Protéines du core viral/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Cellules Sf9 , Antigènes viraux/génétique , Antigènes viraux/immunologie , Lignée cellulaire , Expression des gènes
3.
BMC Infect Dis ; 24(1): 768, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090537

RÉSUMÉ

BACKGROUND: Data on the dynamics and persistence of humoral immunity against SARS-CoV-2 after primary vaccination with two-dose inactivated vaccine (CoronaVac) are limited. This study evaluated the sequential effects of prior infection, heterologous boosting with mRNA-1273 (Moderna), and the occurrence of Omicron vaccine-breakthrough infection (VBI) thereafter. METHODS: We evaluated anti-spike IgG (Abbott) and neutralising (cPASS/GenScript) antibody (nAb) titers up to one year after mRNA-1273 boost in two-dose-CoronaVac-primed Indonesian healthcare workers (August 2021-August 2022). We used linear mixed modeling to estimate the rate of change in antibody levels, and logistic regression to examine associations between antibody levels and VBI. RESULTS: Of 138 participants, 52 (37.7%) had a prior infection and 78 (56.5%) received an mRNA-1273 booster. After two-dose CoronaVac, antibody titers had significantly declined within 180 days, irrespective of prior infection. After mRNA-1273 booster, anti-spike IgG (1.47% decline/day) and Omicron B.1.1.529/BA.2 nAbs declined between day 28-90, and IgG titers plateaued between day 90-360. During the BA.1/BA.2 wave (February-March 2022), 34.6% (27/78) of individuals experienced a VBI (median 181 days after mRNA-1273), although none developed severe illness. VBI was associated with low pre-VBI anti-spike IgG and B.1.1.529/BA.2 nAbs, which were restored post-VBI. CONCLUSIONS: mRNA-1273 booster after two-dose CoronaVac did not prevent BA.1/BA.2 VBI. Periodic vaccine boosters may be warranted against emerging SARS-CoV-2 variants.


Sujet(s)
Vaccin ARNm-1273 contre la COVID-19 , Anticorps neutralisants , Anticorps antiviraux , Réinfections , Vaccins contre la COVID-19 , COVID-19 , Rappel de vaccin , SARS-CoV-2 , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Vaccin ARNm-1273 contre la COVID-19/immunologie , Vaccin ARNm-1273 contre la COVID-19/administration et posologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Réinfections/épidémiologie , Réinfections/immunologie , Réinfections/prévention et contrôle , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/épidémiologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Personnel de santé , Immunoglobuline G/sang , Indonésie/épidémiologie , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Vaccination , Vaccins inactivés/immunologie , Vaccins inactivés/administration et posologie
4.
Front Immunol ; 15: 1442160, 2024.
Article de Anglais | MEDLINE | ID: mdl-39100673

RÉSUMÉ

The COVID-19 pandemic has uncovered the high genetic variability of the SARS-CoV-2 virus and its ability to evade the immune responses that were induced by earlier viral variants. Only a few monoclonal antibodies that have been reported to date are capable of neutralizing a broad spectrum of SARS-CoV-2 variants. Here, we report the isolation of a new broadly neutralizing human monoclonal antibody, iC1. The antibody was identified through sorting the SARS-CoV-1 RBD-stained individual B cells that were isolated from the blood of a vaccinated donor following a breakthrough infection. In vitro, iC1 potently neutralizes pseudoviruses expressing a wide range of SARS-CoV-2 Spike variants, including those of the XBB sublineage. In an hACE2-transgenic mouse model, iC1 provided effective protection against the Wuhan strain of the virus as well as the BA.5 and XBB.1.5 variants. Therefore, iC1 can be considered as a potential component of the broadly neutralizing antibody cocktails resisting the SARS-CoV-2 mutation escape.


Sujet(s)
Angiotensin-converting enzyme 2 , Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Souris transgéniques , SARS-CoV-2 , Animaux , SARS-CoV-2/immunologie , Humains , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Angiotensin-converting enzyme 2/immunologie , Angiotensin-converting enzyme 2/génétique , Angiotensin-converting enzyme 2/métabolisme , Souris , Anticorps antiviraux/immunologie , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Pandémies/prévention et contrôle , Betacoronavirus/immunologie , Betacoronavirus/génétique , Anticorps neutralisants à large spectre/immunologie , Modèles animaux de maladie humaine , Pneumopathie virale/immunologie , Pneumopathie virale/virologie , Pneumopathie virale/prévention et contrôle , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Infections à coronavirus/prévention et contrôle
5.
J Med Virol ; 96(8): e29839, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39105391

RÉSUMÉ

Anti-Spike IgG antibodies against SARS-CoV-2, which are elicited by vaccination and infection, are correlates of protection against infection with pre-Omicron variants. Whether this association can be generalized to infections with Omicron variants is unclear. We conducted a retrospective cohort study with 8457 blood donors in Tyrol, Austria, analyzing 15,340 anti-Spike IgG antibody measurements from March 2021 to December 2022 assessed by Abbott SARS-CoV-2 IgG II chemiluminescent microparticle immunoassay. Using a Bayesian joint model, we estimated antibody trajectories and adjusted hazard ratios for incident SARS-CoV-2 infection ascertained by self-report or seroconversion of anti-Nucleocapsid antibodies. At the time of their earliest available anti-Spike IgG antibody measurement (median November 23, 2021), participants had a median age of 46.0 years (IQR 32.8-55.2), with 45.3% being female, 41.3% having a prior SARS-CoV-2 infection, and 75.5% having received at least one dose of a COVID-19 vaccine. Among 6159 participants with endpoint data, 3700 incident SARS-CoV-2 infections with predominantly Omicron sublineages were recorded over a median of 8.8 months (IQR 5.7-12.4). The age- and sex-adjusted hazard ratio for SARS-CoV-2 associated with having twice the anti-Spike IgG antibody titer was 0.875 (95% credible interval 0.868-0.881) overall, 0.842 (0.827-0.856) during 2021, and 0.884 (0.877-0.891) during 2022 (all p < 0.001). The associations were similar in females and males (Pinteraction = 0.673) and across age (Pinteraction = 0.590). Higher anti-Spike IgG antibody titers were associated with reduced risk of incident SARS-CoV-2 infection across the entire observation period. While the magnitude of association was slightly weakened in the Omicron era, anti-Spike IgG antibody continues to be a suitable correlate of protection against newer SARS-CoV-2 variants.


Sujet(s)
Anticorps antiviraux , COVID-19 , Immunoglobuline G , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Humains , Immunoglobuline G/sang , Mâle , Femelle , SARS-CoV-2/immunologie , Adulte d'âge moyen , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/épidémiologie , Adulte , Études rétrospectives , Glycoprotéine de spicule des coronavirus/immunologie , Autriche/épidémiologie , Vaccins contre la COVID-19/immunologie , Séroconversion , Théorème de Bayes
6.
Bull Exp Biol Med ; 177(2): 221-224, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39093475

RÉSUMÉ

Squalene-based adjuvant compositions that can provide effective induction of specific humoral immune response have been developed. Recombinant receptor-binding domain (RBD) of surface S-protein of SARS-CoV-2 was used to evaluate the properties of the composition. Immunization of mice with the developed squalene-based compositions in combination with RBD allows obtaining high titers of specific antibodies: from 105 to 2×106. The blood sera from immunized mice exhibit neutralizing activity against SARS-CoV-2 Delta variant (B.1.617.2) with a titer up to 1:2000.


Sujet(s)
Adjuvants immunologiques , Anticorps neutralisants , Anticorps antiviraux , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Squalène , Squalène/immunologie , Animaux , Anticorps neutralisants/immunologie , Adjuvants immunologiques/pharmacologie , SARS-CoV-2/immunologie , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Glycoprotéine de spicule des coronavirus/immunologie , COVID-19/immunologie , COVID-19/virologie , Vaccins contre la COVID-19/immunologie , Femelle , Humains , Souris de lignée BALB C , Immunité humorale/effets des médicaments et des substances chimiques
7.
Nat Commun ; 15(1): 6603, 2024 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-39097574

RÉSUMÉ

Vaccine responsiveness is often reduced in older adults. Yet, our lack of understanding of low vaccine responsiveness hampers the development of effective vaccination strategies to reduce the impact of infectious diseases in the ageing population. Young-adult (25-49 y), middle-aged (50-64 y) and older-adult ( ≥ 65 y) participants of the VITAL clinical trials (n = 315, age-range: 28-98 y), were vaccinated with an annual (2019-2020) quadrivalent influenza (QIV) booster vaccine, followed by a primary 13-valent pneumococcal-conjugate (PCV13) vaccine (summer/autumn 2020) and a primary series of two SARS-CoV-2 mRNA-1273 vaccines (spring 2021). This unique setup allowed investigation of humoral responsiveness towards multiple vaccines within the same individuals over the adult age-range. Booster QIV vaccination induced comparable H3N2 hemagglutination inhibition (HI) titers in all age groups, whereas primary PCV13 and mRNA-1273 vaccination induced lower antibody concentrations in older as compared to younger adults (primary endpoint). The persistence of humoral responses, towards the 6 months timepoint, was shorter in older adults for all vaccines (secondary endpoint). Interestingly, highly variable vaccine responder profiles overarching multiple vaccines were observed. Yet, approximately 10% of participants, mainly comprising of older male adults, were classified as low responders to multiple vaccines. This study aids the identification of risk groups for low vaccine responsiveness and hence supports targeted vaccination strategies. Trial number: NL69701.041.19, EudraCT: 2019-000836-24.


Sujet(s)
Vaccin ARNm-1273 contre la COVID-19 , Anticorps antiviraux , COVID-19 , Immunité humorale , Rappel de vaccin , Vaccins antigrippaux , Grippe humaine , Vaccins antipneumococciques , SARS-CoV-2 , Humains , Adulte d'âge moyen , Adulte , Sujet âgé , Mâle , Femelle , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Immunité humorale/immunologie , Vaccins antipneumococciques/immunologie , Vaccins antipneumococciques/administration et posologie , COVID-19/prévention et contrôle , COVID-19/immunologie , SARS-CoV-2/immunologie , Sujet âgé de 80 ans ou plus , Vaccin ARNm-1273 contre la COVID-19/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Facteurs âges , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Sous-type H3N2 du virus de la grippe A/immunologie , Vaccination , Tests d'inhibition de l'hémagglutination
8.
Nat Commun ; 15(1): 6660, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107270

RÉSUMÉ

Safe and effective vaccines against COVID-19 for children and adolescents are needed. This international multicenter, randomized, double-blind, placebo-controlled, phase III clinical trial assessed the efficacy, immunogenicity, and safety of CoronaVac® in children and adolescents (NCT04992260). The study was carried out in Chile, South Africa, Malaysia, and the Philippines. The enrollment ran from September 10, 2021 to March 25, 2022. For efficacy assessment, the median follow-up duration from 14 days after the second dose was 169 days. A total of 11,349 subjects were enrolled. Two 3-µg injections of CoronaVac® or placebo were given 28 days apart. The primary endpoint was the efficacy of the CoronaVac®. The secondary endpoints were the immunogenicity and safety. The vaccine efficacy was 21.02% (95% CI: 1.65, 36.67). The level of neutralizing antibody in the vaccine group was significantly higher than that in the placebo group (GMT: 390.80 vs. 62.20, P <0.0001). Most adverse reactions were mild or moderate. All the severe adverse events were determined to be unrelated to the investigational products. In conclusion, in the Omicron-dominate period, a two-dose schedule of 3 µg CoronaVac® was found to be safe and immunogenic, and showed potential against symptomatic COVID-19 in healthy children and adolescents.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , SARS-CoV-2 , Humains , Adolescent , COVID-19/prévention et contrôle , COVID-19/immunologie , Enfant , Femelle , Mâle , Méthode en double aveugle , Enfant d'âge préscolaire , Nourrisson , SARS-CoV-2/immunologie , Anticorps antiviraux/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/effets indésirables , Vaccins contre la COVID-19/administration et posologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Immunogénicité des vaccins , Philippines , République d'Afrique du Sud , Chili , Malaisie , Vaccins inactivés
9.
Front Immunol ; 15: 1379175, 2024.
Article de Anglais | MEDLINE | ID: mdl-39086481

RÉSUMÉ

Introduction: Intra-tumoral B cells mediate a plethora of immune effector mechanisms with key roles in anti-tumor immunity and serve as positive prognostic indicators in a variety of solid tumor types, including epithelial ovarian cancer (EOC). Several aspects of intra-tumoral B cells remain unclear, such as their state of activation, antigenic repertoires, and capacity to mature into plasma cells. Methods: B lymphocytes were isolated from primary EOC tissue and malignant ascites and were maintained in cell culture medium. The stably maintained cell lines were profiled with flow cytometry and B cell receptor sequencing. Secreted antibodies were tested with a human proteome array comprising more than 21,000 proteins, followed by ELISA for validation. Originating tumor samples were used for spatial profiling with chip cytometry. Results: Antibody-secreting B lymphocytes were isolated from the ovarian tumor microenvironment (TME) of four different EOC patients. The highly clonal cell populations underwent spontaneous immortalization in vitro, were stably maintained in an antibody-secreting state, and showed presence of Epstein-Barr viral (EBV) proteins. All originating tumors had high frequency of tumor-infiltrating B cells, present as lymphoid aggregates, or tertiary lymphoid structures. The antigens recognized by three of the four cell lines are coil-coil domain containing protein 155 (CCDC155), growth factor receptor-bound protein 2 (GRB2), and pyruvate dehydrogenase phosphatase2 (PDP2), respectively. Anti-CCDC155 circulating IgG antibodies were detected in 9 of 20 (45%) of EOC patients' sera. Tissue analyses with multiparameter chip cytometry shows that the antibodies secreted by these novel human B cell lines engage their cognate antigens on tumor cells. Discussion: These studies demonstrate that within the tumor-infiltrating lymphocyte population in EOC resides a low frequency population of antibody-secreting B cells that have been naturally exposed to EBV. Once stably maintained, these novel cell lines offer unique opportunities for future studies on intratumor B cell biology and new target antigen recognition, and for studies on EBV latency and/or viral reactivation in the TME of non-EBV related solid tumors such as the EOC.


Sujet(s)
Ascites , Lymphocytes B , Herpèsvirus humain de type 4 , Tumeurs de l'ovaire , Humains , Femelle , Tumeurs de l'ovaire/immunologie , Herpèsvirus humain de type 4/immunologie , Lymphocytes B/immunologie , Ascites/immunologie , Infections à virus Epstein-Barr/immunologie , Latence virale/immunologie , Microenvironnement tumoral/immunologie , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Carcinome épithélial de l'ovaire/immunologie , Anticorps antiviraux/immunologie , Lignée cellulaire tumorale
10.
PLoS Pathog ; 20(8): e1012393, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39116029

RÉSUMÉ

Live-attenuated influenza vaccines (LAIV) offer advantages over the commonly used inactivated split influenza vaccines. However, finding the optimal balance between sufficient attenuation and immunogenicity has remained a challenge. We recently developed an alternative LAIV based on the 2009 pandemic H1N1 virus with a truncated NS1 protein and lacking PA-X protein expression (NS1(1-126)-ΔPAX). This virus showed a blunted replication and elicited a strong innate immune response. In the present study, we evaluated the efficacy of this vaccine candidate in the porcine animal model as a pertinent in vivo system. Immunization of pigs via the nasal route with the novel NS1(1-126)-ΔPAX LAIV did not cause disease and elicited a strong mucosal immune response that completely blocked replication of the homologous challenge virus in the respiratory tract. However, we observed prolonged shedding of our vaccine candidate from the upper respiratory tract. To improve LAIV safety, we developed a novel prime/boost vaccination strategy combining primary intramuscular immunization with a haemagglutinin-encoding propagation-defective vesicular stomatitis virus (VSV) replicon, followed by a secondary immunization with the NS1(1-126)-ΔPAX LAIV via the nasal route. This two-step immunization procedure significantly reduced LAIV shedding, increased the production of specific serum IgG, neutralizing antibodies, and Th1 memory cells, and resulted in sterilizing immunity against homologous virus challenge. In conclusion, our novel intramuscular prime/intranasal boost regimen interferes with virus shedding and transmission, a feature that will help combat influenza epidemics and pandemics.


Sujet(s)
Administration par voie nasale , Vaccins antigrippaux , Infections à Orthomyxoviridae , Animaux , Suidae , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Injections musculaires , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Sous-type H1N1 du virus de la grippe A/immunologie , Modèles animaux de maladie humaine , Anticorps antiviraux/immunologie , Rappel de vaccin/méthodes , Vaccination/méthodes , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie
11.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125633

RÉSUMÉ

Epstein-Barr virus (EBV) infection and various chemokines, including CCL20, CXCL8 and CXCL10 are considered to participate in the pathogenesis of multiple sclerosis (MS), and several studies point to a direct regulatory effect of EBV on the expression of these chemokines. In our study we hypothesized that serum concentrations of CCL20, CXCL8 and CXCL0 are induced in patients with relapsing-remitting MS (RRMS) in comparison to healthy individuals, and that they are associated with EBV infection. Serum concentrations of CXCL8 and CXCL10 were lower in RRMS patients in relapse in comparison to healthy controls. Although potential effects of corticosteroid therapy introduced in a subgroup of RRMS patients prior to sampling were excluded by subgroup comparison, this possibility has to be considered while interpreting the results. We found an inverse association between serum concentrations of CXCL8 and anti-Epstein-Barr Virus Nuclear Antigen (EBNA) IgG and decreased expression of CXCL8 in peripheral blood mononuclear cells (PBMC) in relapse compared to remission. Lower serum concentrations of CXCL8 and CXCL10 in RRMS patients and decreased peripheral production of CXCL8 in relapse may indicate compensatory anti-inflammatory counter-regulation in MS.


Sujet(s)
Chimiokine CCL20 , Chimiokine CXCL10 , Herpèsvirus humain de type 4 , Interleukine-8 , Sclérose en plaques récurrente-rémittente , Humains , Sclérose en plaques récurrente-rémittente/sang , Sclérose en plaques récurrente-rémittente/immunologie , Sclérose en plaques récurrente-rémittente/traitement médicamenteux , Sclérose en plaques récurrente-rémittente/virologie , Femelle , Chimiokine CXCL10/sang , Adulte , Mâle , Herpèsvirus humain de type 4/immunologie , Chimiokine CCL20/sang , Interleukine-8/sang , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Infections à virus Epstein-Barr/sang , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/virologie , Adulte d'âge moyen , Agranulocytes/métabolisme , Agranulocytes/immunologie , Études cas-témoins
12.
Virol J ; 21(1): 177, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107806

RÉSUMÉ

BACKGROUND: Reticuloendotheliosis virus (REV), a member of the family Retroviridae, is a hot area of research, and a previous study showed that exosomes purified from REV-positive semen were not blocked by REV-specific neutralizing antibodies and established productive infections. METHODS: To further verify the infectivity of exosomes from REV-infected cells, we isolated and purified exosomes from REV-infected DF-1 cells and identified them using Western blot and a transmission electron microscope. We then inoculated 7-day-old embryonated eggs, 1-day-old chicks and 23-week-old hens with and without antibody treatment. REV was administered simultaneously as a control. RESULTS: In the absence of antibodies, the results indicated that REV-exosomes and REV could infect chicks, resulting in viremia and viral shedding, compared with the infection caused by REV, REV-exosomes reduced the hatching rate and increased mortality after hatching, causing severe growth inhibition and immune organ damage in 1-day-old chicks; both REV and REV-exosomes also could infect hens, however, lead to transient infection. In the presence of antibodies, REV-exosomes were not blocked by REV-specific neutralizing antibodies and infected 7-day-old embryonated eggs. However, REV could not infect 1-day-old chicks and 23-week-old hens. CONCLUSION: In this study, we compared the infectious ability of REV-exosomes and REV, REV-exosomes could escape from REV-specific neutralizing antibodies in embryonated eggs, providing new insights into the immune escape mechanism of REV.


Sujet(s)
Anticorps antiviraux , Poulets , Exosomes , Maladies de la volaille , Virus de la réticuloendothéliose , Infections à Retroviridae , Excrétion virale , Animaux , Exosomes/virologie , Exosomes/immunologie , Anticorps antiviraux/immunologie , Poulets/virologie , Virus de la réticuloendothéliose/immunologie , Maladies de la volaille/virologie , Maladies de la volaille/transmission , Maladies de la volaille/immunologie , Infections à Retroviridae/virologie , Infections à Retroviridae/transmission , Infections à Retroviridae/immunologie , Infections à Retroviridae/médecine vétérinaire , Anticorps neutralisants/immunologie , Lignée cellulaire , Virémie/virologie , Femelle
13.
Hum Vaccin Immunother ; 20(1): 2384192, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39149872

RÉSUMÉ

Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.


Sujet(s)
Anticorps antiviraux , Mémoire immunologique , Humains , Anticorps antiviraux/immunologie , Animaux , Vaccins antiviraux/immunologie
14.
Nat Commun ; 15(1): 7009, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39147753

RÉSUMÉ

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


Sujet(s)
Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , Épitopes , Phlebovirus , Syndrome de fièvre sévère avec thrombocytopénie , Animaux , Phlebovirus/immunologie , Souris , Anticorps neutralisants/immunologie , Anticorps neutralisants/usage thérapeutique , Anticorps monoclonaux/immunologie , Anticorps antiviraux/immunologie , Syndrome de fièvre sévère avec thrombocytopénie/immunologie , Syndrome de fièvre sévère avec thrombocytopénie/virologie , Syndrome de fièvre sévère avec thrombocytopénie/prévention et contrôle , Humains , Épitopes/immunologie , Femelle , Souris de lignée BALB C , Protéines de l'enveloppe virale/immunologie , Cristallographie aux rayons X , Chlorocebus aethiops , Glycoprotéines/immunologie , Cellules Vero
16.
Nat Commun ; 15(1): 7033, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39147778

RÉSUMÉ

The SARS-CoV-2 Omicron variant sparked the largest wave of infections worldwide. Mainland China eased its strict COVID-19 measures in late 2022 and experienced two nationwide Omicron waves in 2023. Here, we investigated lineage distribution and virus evolution in Guangdong, China, 2022-2023 by comparing 5813 local viral genomes with the datasets from other regions of China and worldwide. Additionally, we conducted three large-scale serological surveys involving 1696 participants to measure their immune response to the BA.5 and XBB.1.9 before and after the corresponding waves. Our findings revealed the Omicron variants, mainly the BA.5.2.48 lineage, causing infections in over 90% of individuals across different age groups within a month. This rapid spread led to the establishment of widespread immunity, limiting the virus's ability to further adaptive mutation and dissemination. While similar immune responses to BA.5 were observed across all age groups after the initial wave, children aged 3 to 11 developed a stronger cross immune response to the XBB.1.9 strain, possibly explaining their lower infection rates in the following XBB.1 wave. Reinfection with Omicron XBB.1 variant triggered a more potent neutralizing immune response among older adults. These findings highlight the impact of age-specific immune responses on viral spread in potential future waves.


Sujet(s)
COVID-19 , Génome viral , SARS-CoV-2 , Humains , COVID-19/immunologie , COVID-19/épidémiologie , COVID-19/virologie , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Chine/épidémiologie , Enfant , Enfant d'âge préscolaire , Adulte , Adolescent , Adulte d'âge moyen , Jeune adulte , Génome viral/génétique , Mâle , Femelle , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Épidémiologie moléculaire , Nourrisson , Sujet âgé , Pandémies , Phylogenèse
17.
Front Immunol ; 15: 1383086, 2024.
Article de Anglais | MEDLINE | ID: mdl-39119342

RÉSUMÉ

Current vaccines against COVID-19 elicit immune responses that are overall strong but wane rapidly. As a consequence, the necessary booster shots have contributed to vaccine fatigue. Hence, vaccines that would provide lasting protection against COVID-19 are needed, but are still unavailable. Cytomegaloviruses (CMVs) elicit lasting and uniquely strong immune responses. Used as vaccine vectors, they may be attractive tools that obviate the need for boosters. Therefore, we tested the murine CMV (MCMV) as a vaccine vector against COVID-19 in relevant preclinical models of immunization and challenge. We have previously developed a recombinant MCMV vaccine vector expressing the spike protein of the ancestral SARS-CoV-2 (MCMVS). In this study, we show that the MCMVS elicits a robust and lasting protection in young and aged mice. Notably, spike-specific humoral and cellular immunity was not only maintained but also even increased over a period of at least 6 months. During that time, antibody avidity continuously increased and expanded in breadth, resulting in neutralization of genetically distant variants, like Omicron BA.1. A single dose of MCMVS conferred rapid virus clearance upon challenge. Moreover, MCMVS vaccination controlled two variants of concern (VOCs), the Beta (B.1.135) and the Omicron (BA.1) variants. Thus, CMV vectors provide unique advantages over other vaccine technologies, eliciting broadly reactive and long-lasting immune responses against COVID-19.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Souris , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Muromegalovirus/immunologie , Muromegalovirus/génétique , Femelle , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Souris de lignée BALB C , Humains , Vecteurs génétiques , Immunité cellulaire , Immunité humorale , Modèles animaux de maladie humaine
18.
Front Immunol ; 15: 1385121, 2024.
Article de Anglais | MEDLINE | ID: mdl-39119337

RÉSUMÉ

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


Sujet(s)
Anticorps antiviraux , Immunoglobuline G , Lait humain , Rhinovirus , Humains , Lait humain/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Femelle , Immunoglobuline G/immunologie , Immunoglobuline G/sang , Nourrisson , Rhinovirus/immunologie , Immunoglobuline A/immunologie , Immunoglobuline A/sang , Infections à Picornaviridae/immunologie , Nouveau-né , Épitopes/immunologie , Protéines de capside/immunologie , Adulte
19.
Front Immunol ; 15: 1416375, 2024.
Article de Anglais | MEDLINE | ID: mdl-39131158

RÉSUMÉ

With the rapid global spread of COVID-19 and the continuous emergence of variants, there is an urgent need to develop safe and effective vaccines. Here, we developed a novel mRNA vaccine, HC009, based on new formulation by the QTsome delivery platform. Immunogenicity results showed that the prime-boost immunization strategy with HC009 was able to induce robust and durable humoral immunity, as well as Th1-biased cellular responses in rodents or non-human primates (NHPs). After further challenge with live SARS-CoV-2 virus, HC009 provided adequate protection against virus infection in hACE2 transgenic mice. Therefore, HC009 could provide significant immune protection against SARS-CoV-2.


Sujet(s)
Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Immunogénicité des vaccins , Souris transgéniques , SARS-CoV-2 , Vaccins à ARNm , Animaux , SARS-CoV-2/immunologie , Vaccins contre la COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Souris , Vaccins à ARNm/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Humains , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Immunité humorale , Femelle , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Souris de lignée BALB C ,
20.
Sci Transl Med ; 16(759): eadi1625, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39110777

RÉSUMÉ

The recent emergence and rapid response to severe acute respiratory syndrome coronavirus 2 was enabled by prototype pathogen and vaccine platform approaches, driven by the preemptive application of RNA vaccine technology to the related Middle East respiratory syndrome coronavirus. Recently, the National Institutes of Allergy and Infectious Diseases identified nine virus families of concern, eight enveloped virus families and one nonenveloped virus family, for which vaccine generation is a priority. Although RNA vaccines have been described for a variety of enveloped viruses, a roadmap for their use against nonenveloped viruses is lacking. Enterovirus D68 was recently designated a prototype pathogen within the family Picornaviridae of nonenveloped viruses because of its rapid evolution and respiratory route of transmission, coupled with a lack of diverse anti-enterovirus vaccine approaches in development. Here, we describe a proof-of-concept approach using a clinical stage RNA vaccine platform that induced robust enterovirus D68-neutralizing antibody responses in mice and nonhuman primates and prevented upper and lower respiratory tract infections and neurological disease in mice. In addition, we used our platform to rapidly characterize the antigenic diversity within the six genotypes of enterovirus D68, providing the necessary data to inform multivalent vaccine compositions that can elicit optimal breadth of neutralizing responses. These results demonstrate that RNA vaccines can be used as tools in our pandemic-preparedness toolbox for nonenveloped viruses.


Sujet(s)
Anticorps neutralisants , Entérovirus humain D , Infections à entérovirus , Animaux , Infections à entérovirus/prévention et contrôle , Infections à entérovirus/immunologie , Infections à entérovirus/virologie , Entérovirus humain D/immunologie , Entérovirus humain D/génétique , Anticorps neutralisants/immunologie , Souris , Vaccins antiviraux/immunologie , Modèles animaux de maladie humaine , Humains , Vaccins à ARNm , Anticorps antiviraux/immunologie , Femelle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE