Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24.937
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000308

RÉSUMÉ

Viral infection generally induces polyclonal neutralizing antibody responses. However, how many lineages of antibody responses can fully represent the neutralization activities in sera has not been well studied. Using the newly designed stable HIV-1 Env trimer as hook, we isolated two distinct broadly neutralizing antibodies (bnAbs) from Chinese rhesus macaques infected with SHIV1157ipd3N4 for 5 years. One lineage of neutralizing antibodies (JT15 and JT16) targeted the V2-apex in the Env trimers, similar to the J038 lineage bnAbs identified in our previous study. The other lineage neutralizing antibody (JT18) targeted the V3 crown region in the Env, which strongly competed with human 447-52D. Each lineage antibody neutralized a different set of viruses. Interestingly, when the two neutralizing antibodies from different lineages isolated from the same macaque were combined, the mixture had a neutralization breath very similar to that from the cognate sera. Our study demonstrated that a minimum of two different neutralizing antibodies can fully recapitulate the serum neutralization breadth. This observation can have important implications in AIDS vaccine design.


Sujet(s)
Anticorps neutralisants , Anticorps anti-VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Macaca mulatta , Syndrome d'immunodéficience acquise du singe , Macaca mulatta/immunologie , Animaux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/sang , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Humains , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/sang , Syndrome d'immunodéficience acquise du singe/virologie , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/sang , Virus de l'immunodéficience simienne/immunologie , Produits du gène env du virus de l'immunodéficience humaine/immunologie , Tests de neutralisation
2.
Proc Natl Acad Sci U S A ; 121(29): e2310421121, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38976733

RÉSUMÉ

We generated a replication-competent OC43 human seasonal coronavirus (CoV) expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike in place of the native spike (rOC43-CoV2 S). This virus is highly attenuated relative to OC43 and SARS-CoV-2 in cultured cells and animals and is classified as a biosafety level 2 (BSL-2) agent by the NIH biosafety committee. Neutralization of rOC43-CoV2 S and SARS-CoV-2 by S-specific monoclonal antibodies and human sera is highly correlated, unlike recombinant vesicular stomatitis virus-CoV2 S. Single-dose immunization with rOC43-CoV2 S generates high levels of neutralizing antibodies against SARS-CoV-2 and fully protects human ACE2 transgenic mice from SARS-CoV-2 lethal challenge, despite nondetectable replication in respiratory and nonrespiratory organs. rOC43-CoV2 S induces S-specific serum and airway mucosal immunoglobulin A and IgG responses in rhesus macaques. rOC43-CoV2 S has enormous value as a BSL-2 agent to measure S-specific antibodies in the context of a bona fide CoV and is a candidate live attenuated SARS-CoV-2 mucosal vaccine that preferentially replicates in the upper airway.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Tests de neutralisation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Humains , Anticorps neutralisants/immunologie , Souris , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Anticorps antiviraux/immunologie , Tests de neutralisation/méthodes , Souris transgéniques , Coronavirus humain OC43/immunologie , Coronavirus humain OC43/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/immunologie , Chlorocebus aethiops , Cellules Vero , Macaca mulatta
3.
J Med Virol ; 96(7): e29801, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38988204

RÉSUMÉ

SARS-CoV-2 Omicron lineages continue to emerge and evolve into new sublineages, causing infection waves throughout 2022 and 2023, which has been attributed to immune escape. We examined neutralizing antibody responses to the recently emerged SARS-CoV-2 JN.1 variant in comparison to ancestral D614G and Omicron BA.1, BA.2, BA.5, and XBB.1.5 variants. We tested 79 human sera from cohorts with different combinations of vaccinations and infections, including 23 individuals who had been repeatedly exposed to Omicron. Individuals with a monovalent XBB.1.5 vaccine booster or XBB.1.5 breakthrough infection had robust antibody levels against all variants tested; however, JN.1 evaded antibodies in individuals after single Omicron BA.1, BA.2 or BA.5 breakthrough infections. Moreover, in the non-vaccinated cohort, serum antibodies demonstrated almost no cross-neutralization activities against D614G, XBB.1.5 and JN.1. after infections with earlier Omicron variants. These findings show that SARS-CoV-2-immunity is heterogeneous, depending on different combinations of vaccinations and infections, and emphasize the importance of considering different immune-backgrounds when evaluating novel variants.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Rappel de vaccin , SARS-CoV-2 , Humains , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , COVID-19/immunologie , COVID-19/prévention et contrôle , COVID-19/virologie , Anticorps antiviraux/sang , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Femelle , Mâle , Adulte , Adulte d'âge moyen , Vaccination , Tests de neutralisation , Sujet âgé
4.
Vet Microbiol ; 295: 110167, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38954881

RÉSUMÉ

Hendra virus (HeV) is lethal to horses and a zoonotic threat to humans in Australia, causing severe neurological and/or respiratory disease with high mortality. An equine vaccine has been available since 2012. Foals acquire antibodies from their dams by ingesting colostrum after parturition, therefore it is assumed that foals of mares vaccinated against HeV will have passive HeV antibodies circulating during the first several months of life until they are actively vaccinated. However, no studies have yet examined passive or active immunity against HeV in foals. Here, we investigated anti-HeV antibody levels in vaccinated mares and their foals. Testing for HeV neutralising antibodies is cumbersome due to the requirement for Biosafety level 4 (BSL-4) containment to conduct virus neutralisation tests (VNT). For this study, a subset of samples was tested for HeV G-specific antibodies by both an authentic VNT with infectious HeV and a microsphere-based immunoassay (MIA), revealing a strong correlation. An indicative neutralising level was then applied to the results of a larger sample set tested using the MIA. Mares had high levels of HeV-specific neutralising antibodies at the time of parturition. Foals acquired high levels of maternal antibodies which then waned to below predictive protective levels in most foals by 6 months old when vaccination commenced. Foals showed a suboptimal response to vaccination, suggesting maternal antibodies may interfere with active vaccination. The correlation analysis between the authentic HeV VNT and HeV MIA will enable further high throughput serological studies to inform optimal vaccination protocols for both broodmares and foals.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Virus Hendra , Infections à hénipavirus , Maladies des chevaux , Vaccination , Vaccins antiviraux , Animaux , Equus caballus , Virus Hendra/immunologie , Maladies des chevaux/prévention et contrôle , Maladies des chevaux/virologie , Maladies des chevaux/immunologie , Anticorps antiviraux/sang , Infections à hénipavirus/prévention et contrôle , Infections à hénipavirus/médecine vétérinaire , Infections à hénipavirus/immunologie , Infections à hénipavirus/virologie , Femelle , Vaccination/médecine vétérinaire , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Anticorps neutralisants/sang , Immunité acquise d'origine maternelle , Animaux nouveau-nés/immunologie , Grossesse , Tests de neutralisation/médecine vétérinaire , Australie , Colostrum/immunologie
5.
Emerg Microbes Infect ; 13(1): 2373307, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38953857

RÉSUMÉ

SARS-CoV-2 has been evolving into a large number of variants, including the highly pathogenic Delta variant, and the currently prevalent Omicron subvariants with extensive evasion capability, which raises an urgent need to develop new broad-spectrum neutralizing antibodies. Herein, we engineer two IgG-(scFv)2 form bispecific antibodies with overlapping epitopes (bsAb1) or non-overlapping epitopes (bsAb2). Both bsAbs are significantly superior to the parental monoclonal antibodies in terms of their antigen-binding and virus-neutralizing activities against all tested circulating SARS-CoV-2 variants including currently dominant JN.1. The bsAb1 can efficiently neutralize all variants insensitive to parental monoclonal antibodies or the cocktail with IC50 lower than 20 ng/mL, even slightly better than bsAb2. Furthermore, the cryo-EM structures of bsAb1 in complex with the Omicron spike protein revealed that bsAb1 with overlapping epitopes effectively locked the S protein, which accounts for its conserved neutralization against Omicron variants. The bispecific antibody strategy engineered from overlapping epitopes provides a novel solution for dealing with viral immune evasion.


Sujet(s)
Anticorps bispécifiques , Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Épitopes , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/composition chimique , Anticorps bispécifiques/pharmacologie , SARS-CoV-2/immunologie , Anticorps neutralisants/immunologie , Humains , Anticorps antiviraux/immunologie , Épitopes/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/composition chimique , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Tests de neutralisation
6.
BMC Immunol ; 25(1): 43, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987686

RÉSUMÉ

OBJECTIVE: HIV has been reported to interfere with protective vaccination against multiple pathogens, usually through the decreased effectiveness of the antibody responses. We aimed to assess neutralizing antibody responses induced by COVID-19 vaccination in PLWH in Brazzaville, Republique of the Congo. METHOD: The study was conducted at the Ambulatory Treatment Center of the National HIV Program, in charge of over 6000 PLWH, and the health center of FCRM in Brazzaville, Republic of the Congo. Participants were divided into two groups: PLWH with well-controlled HIV infection (CD4 counts no older than one week ≥ 800 / mm3, undetectable viral load of a period no older than one week and regularly taking Highly Active Antiretroviral Therapy for at least 6 months) and PLWOH. These groups were subdivided by vaccination status: fully vaccinated with adenovirus-based vaccines (Janssen/Ad26.COV2.S and Sputnik/Gam-COVID-Vac) or inactivated virus vaccine (Sinopharm/BBIP-CorV) and a control group of unvaccinated healthy individuals. All participants were RT-PCR negative at inclusion and/or with no documented history of SARS-CoV-2 infection. ELISA method was used for detecting IgG and neutralizing Antibodies against SARS-CoV-2 antigens using a commercial neutralizing assay. RESULTS: We collected oropharyngeal and blood samples from 1016 participants including 684 PLWH and 332 PLWOH. Both PLWH and PLWOH elicited high levels of antibody responses after complete vaccination with inactivated virus vaccine (Sinopharm/BBIP-CorV) and adenovirus-based vaccines (Janssen/Ad26.COV2.S and Sputnik/Gam-COVID-Vac). Overall, no difference was observed in neutralization capacity between PLWOH and PLWH with well-controlled HIV infection. CONCLUSION: The results from this study underline the importance of implementing integrated health systems that provide PLWH the opportunity to benefit HIV prevention and care, at the same time while monitoring their vaccine-induced antibody kinetics for appropriate booster schedules.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre la COVID-19 , COVID-19 , Infections à VIH , SARS-CoV-2 , Vaccination , Humains , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Infections à VIH/immunologie , Infections à VIH/traitement médicamenteux , Mâle , Femelle , COVID-19/immunologie , COVID-19/prévention et contrôle , Vaccins contre la COVID-19/immunologie , Adulte , SARS-CoV-2/immunologie , Adulte d'âge moyen , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Tests de neutralisation
7.
Virol J ; 21(1): 148, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951814

RÉSUMÉ

The magnitude of the HIV-1 epidemic in Nigeria is second only to the subtype C epidemic in South Africa, yet the subtypes prevalent in Nigeria require further characterization. A panel of 50 subtype G and 18 CRF02_AG Nigerian HIV-1 pseudoviruses (PSV) was developed and envelope coreceptor usage, neutralization sensitivity and cross-clade reactivity were characterized. These PSV were neutralized by some antibodies targeting major neutralizing determinants, but potentially important differences were observed in specific sensitivities (eg. to sCD4, MPER and V2/V3 monoclonal antibodies), as well as in properties such as variable loop lengths, number of potential N-linked glycans and charge, demonstrating distinct antigenic characteristics of CRF02_AG and subtype G. There was preferential neutralization of the matched CRF/subtype when PSV from subtype G or CRF02_AG were tested using pooled plasma. These novel Nigerian PSV will be useful to study HIV-1 CRF- or subtype-specific humoral immune responses for subtype G and CRF02_AG.


Sujet(s)
Anticorps neutralisants , Anticorps anti-VIH , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Tests de neutralisation , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/classification , Nigeria , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Humains , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/sang , Infections à VIH/immunologie , Infections à VIH/virologie , Produits du gène env du virus de l'immunodéficience humaine/immunologie , Produits du gène env du virus de l'immunodéficience humaine/génétique , Réactions croisées/immunologie
8.
J Med Microbiol ; 73(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38963412

RÉSUMÉ

Introduction. In India, the SARS-CoV-2 Delta wave (2020-2021) faded away with the advent of the Omicron variants (2021-present). Dengue incidences were observed to be less in Southeast Asia during the active years of the pandemic (2020-2021). However, dengue virus type 3 (DV3) cases were increasingly reported in this region (including India) concurrent with the progression of the Omicron waves since 2022.Hypothesis. What could be the reason(s) behind this unusual DV3 surge after an overall dip in dengue incidences in many parts of Southeast Asia?Aim. We, therefore, investigated the current state of cross-reactivity of prevalent (Omicron era) SARS-CoV-2 serums with different DV serotypes and evaluated the impact of such serums on DV neutralization in cell culture.Methodology. Fifty-five COVID-19 serum samples (January-September 2022) and three pre-pandemic archived serum samples from apparently healthy individuals were tested for DV or SARS-CoV-2 IgM/IgG using the lateral flow immunoassays. DV1-4 virus neutralization tests (VNTs) were done with the SARS-CoV-2 antibody (Ab)-positive serums in Huh7 cells. DV3 envelope (env) gene was PCR amplified and sequenced for three archived DV isolates, one from 2017 and two from 2021.Results. SARS-CoV-2 Ab-positive samples constituted 74.5 % of the serums. Of these, 41.5 % were DV cross-reactive and 58.5 % were not. The DV cross-reactive serums neutralized all DV serotypes (DV1-4), as per previous results and this study. The DV non-cross-reactive serums (58.5 %) also cross-neutralized DV1, 2 and 4 but increased DV3 infectivity by means of antibody-dependent enhancement of infection as evident from significantly higher DV3 titres in VNT compared to control serums. The DV3 envelope was identical among the three isolates, including isolate 1 used in VNTs. Our results suggest that DV cross-reactivity of SARS-CoV-2 serums diminished with the shift from Delta to Omicron prevalence. Such COVID-19 serums (DV non-cross-reactive) might have played a major role in causing DV3 surge during the Omicron waves.Conclusion. Patients suspected of dengue or COVID-19 should be subjected to virus/antigen tests and serological tests for both the diseases for definitive diagnosis, prognosis and disease management.


Sujet(s)
Anticorps antiviraux , COVID-19 , Réactions croisées , Virus de la dengue , SARS-CoV-2 , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , COVID-19/virologie , COVID-19/épidémiologie , COVID-19/sang , COVID-19/immunologie , Anticorps antiviraux/sang , Virus de la dengue/génétique , Virus de la dengue/immunologie , Virus de la dengue/classification , Inde/épidémiologie , Dengue/virologie , Dengue/sang , Dengue/épidémiologie , Dengue/immunologie , Tests de neutralisation , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Immunoglobuline G/sang , Immunoglobuline M/sang
9.
Nature ; 631(8021): 617-626, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38961298

RÉSUMÉ

SARS-CoV-2 variants acquire mutations in the spike protein that promote immune evasion1 and affect other properties that contribute to viral fitness, such as ACE2 receptor binding and cell entry2,3. Knowledge of how mutations affect these spike phenotypes can provide insight into the current and potential future evolution of the virus. Here we use pseudovirus deep mutational scanning4 to measure how more than 9,000 mutations across the full XBB.1.5 and BA.2 spikes affect ACE2 binding, cell entry or escape from human sera. We find that mutations outside the receptor-binding domain (RBD) have meaningfully affected ACE2 binding during SARS-CoV-2 evolution. We also measure how mutations to the XBB.1.5 spike affect neutralization by serum from individuals who recently had SARS-CoV-2 infections. The strongest serum escape mutations are in the RBD at sites 357, 420, 440, 456 and 473; however, the antigenic effects of these mutations vary across individuals. We also identify strong escape mutations outside the RBD; however, many of them decrease ACE2 binding, suggesting they act by modulating RBD conformation. Notably, the growth rates of human SARS-CoV-2 clades can be explained in substantial part by the measured effects of mutations on spike phenotypes, suggesting our data could enable better prediction of viral evolution.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Mutation , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/métabolisme , Glycoprotéine de spicule des coronavirus/immunologie , Humains , SARS-CoV-2/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/classification , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , COVID-19/virologie , COVID-19/immunologie , COVID-19/génétique , Liaison aux protéines , Échappement immunitaire/génétique , Anticorps neutralisants/immunologie , Pénétration virale , Évolution moléculaire , Modèles moléculaires , Sites de fixation , Domaines protéiques , Tests de neutralisation , Analyse de mutations d'ADN
10.
J Gen Virol ; 105(6)2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38861287

RÉSUMÉ

Increased human-to-human transmission of monkeypox virus (MPXV) is cause for concern, and antibodies directed against vaccinia virus (VACV) are known to confer cross-protection against Mpox. We used 430 serum samples derived from the Scottish patient population to investigate antibody-mediated cross-neutralization against MPXV. By combining electrochemiluminescence immunoassays with live-virus neutralization assays, we show that people born when smallpox vaccination was routinely offered in the United Kingdom have increased levels of antibodies that cross-neutralize MPXV. Our results suggest that age is a risk factor of Mpox infection, and people born after 1971 are at higher risk of infection upon exposure.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Virus de la variole simienne , Orthopoxvirose simienne , Vaccin antivariolique , Humains , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccin antivariolique/immunologie , Vaccin antivariolique/administration et posologie , Adulte , Adulte d'âge moyen , Virus de la variole simienne/immunologie , Jeune adulte , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Orthopoxvirose simienne/immunologie , Orthopoxvirose simienne/prévention et contrôle , Femelle , Adolescent , Sujet âgé , Mâle , Protection croisée/immunologie , Écosse , Facteurs âges , Tests de neutralisation , Enfant , Vaccination , Variole/prévention et contrôle , Variole/immunologie , Enfant d'âge préscolaire , Réactions croisées , Sujet âgé de 80 ans ou plus
11.
J Med Virol ; 96(6): e29743, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38884419

RÉSUMÉ

As one of the most effective measures to prevent seasonal influenza viruses, annual influenza vaccination is globally recommended. Nevertheless, evidence regarding the impact of repeated vaccination to contemporary and future influenza has been inconclusive. A total of 100 subjects singly or repeatedly immunized with influenza vaccines including 3C.2a1 or 3C.3a1 A(H3N2) during 2018-2019 and 2019-2020 influenza season were recruited. We investigated neutralization antibody by microneutralization assay using four antigenically distinct A(H3N2) viruses circulating from 2018 to 2023, and tracked the dynamics of B cell receptor (BCR) repertoire for consecutive vaccinations. We found that vaccination elicited cross-reactive antibody responses against future emerging strains. Broader neutralizing antibodies to A(H3N2) viruses and more diverse BCR repertoires were observed in the repeated vaccination. Meanwhile, a higher frequency of BCR sequences shared among the repeated-vaccinated individuals with consistently boosting antibody response was found than those with a reduced antibody response. Our findings suggest that repeated seasonal vaccination could broaden the breadth of antibody responses, which may improve vaccine protection against future emerging viruses.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Réactions croisées , Sous-type H3N2 du virus de la grippe A , Vaccins antigrippaux , Grippe humaine , Humains , Sous-type H3N2 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Adulte , Réactions croisées/immunologie , Mâle , Femelle , Vaccination , Adulte d'âge moyen , Jeune adulte , Tests de neutralisation , Récepteurs pour l'antigène des lymphocytes B/immunologie , Récepteurs pour l'antigène des lymphocytes B/génétique , Adolescent
12.
Cell Mol Life Sci ; 81(1): 267, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884678

RÉSUMÉ

Neutralizing antibodies are considered a correlate of protection against severe human respiratory syncytial virus (HRSV) disease. Currently, HRSV neutralization assays are performed on immortalized cell lines like Vero or A549 cells. It is known that assays on these cell lines exclusively detect neutralizing antibodies (nAbs) directed to the fusion (F) protein. For the detection of nAbs directed to the glycoprotein (G), ciliated epithelial cells expressing the cellular receptor CX3CR1 are required, but generation of primary cell cultures is expensive and labor-intensive. Here, we developed a high-throughput neutralization assay based on the interaction between clinically relevant HRSV grown on primary cells with ciliated epithelial cells, and validated this assay using a panel of infant sera. To develop the high-throughput neutralization assay, we established a culture of differentiated apical-out airway organoids (Ap-O AO). CX3CR1 expression was confirmed, and both F- and G-specific monoclonal antibodies neutralized HRSV in the Ap-O AO. In a side-by-side neutralization assay on Vero cells and Ap-O AO, neutralizing antibody levels in sera from 125 infants correlated well, although titers on Ap-O AO were consistently lower. We speculate that these lower titers might be an actual reflection of the neutralizing antibody capacity in vivo. The organoid-based neutralization assay described here holds promise for further characterization of correlates of protection against HRSV disease.


Sujet(s)
Anticorps neutralisants , Récepteur-1 de la chimiokine CX3C , Tests de neutralisation , Organoïdes , Infections à virus respiratoire syncytial , Virus respiratoire syncytial humain , Humains , Virus respiratoire syncytial humain/immunologie , Anticorps neutralisants/immunologie , Organoïdes/métabolisme , Organoïdes/immunologie , Organoïdes/virologie , Organoïdes/cytologie , Animaux , Tests de neutralisation/méthodes , Chlorocebus aethiops , Cellules Vero , Infections à virus respiratoire syncytial/immunologie , Infections à virus respiratoire syncytial/virologie , Récepteur-1 de la chimiokine CX3C/métabolisme , Récepteur-1 de la chimiokine CX3C/immunologie , Anticorps antiviraux/immunologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/métabolisme , Nourrisson , Cellules épithéliales/métabolisme , Cellules épithéliales/immunologie , Cellules épithéliales/virologie , Anticorps monoclonaux/immunologie
13.
Vet Microbiol ; 295: 110123, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38889619

RÉSUMÉ

Akabane virus (AKAV) is characterized by abortion, stillbirth, premature birth, and congenital deformities in livestock and is widely distributed throughout Australia, Southeast Asia, East Asia, the Middle East, and Africa. Gc protein is the major neutralizing target of AKAV and is often considered as an immunogen to prepare neutralizing antibodies. In this study, we prepared and characterized three monoclonal antibodies (mAbs), 4D1, 4E6, and 4F12, against the Gc protein of AKAV (TJ2016 strain). Western blot (WB) and indirect immunofluorescence assay (IFA) analysis proved that the mAbs can react with both the truncated recombinant AKAV Gc protein and the natural Gc protein produced in the AKAV-infected cells. Further research demonstrated that these mAbs possess neutralizing activity. We next defined a neutralizing epitope 1134SVQSFDGKL1142 by screening a panel of overlapping peptides spanning the truncated Gc protein (aa991∼1232) using the generated neutralizing mAbs. Bioinformatic analysis shows that the neutralizing epitope is highly conserved across different genotypes of AKAV. The newly produced neutralizing mAbs and the identified neutralizing epitope in this study enrich the antigenic epitope information of the AKAV Gc protein and could have potential applications in the development of antigen and antibody detection systems that are specific to AKAV.


Sujet(s)
Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , Épitopes , Orthobunyavirus , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Animaux , Épitopes/immunologie , Anticorps antiviraux/immunologie , Orthobunyavirus/immunologie , Souris , Souris de lignée BALB C , Tests de neutralisation
14.
J Immunol Methods ; 530: 113698, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823574

RÉSUMÉ

There is a critical need to understand the effectiveness of serum elicited by different SARS-CoV-2 vaccines against SARS-CoV-2 variants. We describe the generation of reference reagents comprised of post-vaccination sera from recipients of different primary vaccines with or without different vaccine booster regimens in order to allow standardized characterization of SARS-CoV-2 neutralization in vitro. We prepared and pooled serum obtained from donors who received a either primary vaccine series alone, or a vaccination strategy that included primary and boosted immunization using available SARS-CoV-2 mRNA vaccines (BNT162b2, Pfizer and mRNA-1273, Moderna), replication-incompetent adenovirus type 26 vaccine (Ad26.COV2·S, Johnson and Johnson), or recombinant baculovirus-expressed spike protein in a nanoparticle vaccine plus Matrix-M adjuvant (NVX-CoV2373, Novavax). No subjects had a history of clinical SARS-CoV-2 infection, and sera were screened with confirmation that there were no nucleocapsid antibodies detected to suggest natural infection. Twice frozen sera were aliquoted, and serum antibodies were characterized for SARS-CoV-2 spike protein binding (estimated WHO antibody binding units/ml), spike protein competition for ACE-2 binding, and SARS-CoV-2 spike protein pseudotyped lentivirus transduction. These reagents are available for distribution to the research community (BEI Resources), and should allow the direct comparison of antibody neutralization results between different laboratories. Further, these sera are an important tool to evaluate the functional neutralization activity of vaccine-induced antibodies against emerging SARS-CoV-2 variants of concern. IMPORTANCE: The explosion of COVID-19 demonstrated how novel coronaviruses can rapidly spread and evolve following introduction into human hosts. The extent of vaccine- and infection-induced protection against infection and disease severity is reduced over time due to the fall in concentration, and due to emerging variants that have altered antibody binding regions on the viral envelope spike protein. Here, we pooled sera obtained from individuals who were immunized with different SARS-CoV-2 vaccines and who did not have clinical or serologic evidence of prior infection. The sera pools were characterized for direct spike protein binding, blockade of virus-receptor binding, and neutralization of spike protein pseudotyped lentiviruses. These sera pools were aliquoted and are available to allow inter-laboratory comparison of results and to provide a tool to determine the effectiveness of prior vaccines in recognizing and neutralizing emerging variants of concern.


Sujet(s)
Vaccin ARNm-1273 contre la COVID-19 , Anticorps neutralisants , Anticorps antiviraux , Vaccin BNT162 , Vaccins contre la COVID-19 , COVID-19 , Tests de neutralisation , SARS-CoV-2 , Humains , SARS-CoV-2/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , COVID-19/virologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Vaccin ARNm-1273 contre la COVID-19/immunologie , Vaccin BNT162/immunologie , Vaccin BNT162/administration et posologie , Glycoprotéine de spicule des coronavirus/immunologie , Normes de référence , Rappel de vaccin , Vaccination , Ad26COVS1/immunologie
15.
Nat Commun ; 15(1): 5127, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879565

RÉSUMÉ

The Omicron subvariants BQ.1.1, XBB.1.5, and XBB.1.16 of SARS-CoV-2 are known for their adeptness at evading immune responses. Here, we isolate a neutralizing antibody, 7F3, with the capacity to neutralize all tested SARS-CoV-2 variants, including BQ.1.1, XBB.1.5, and XBB.1.16. 7F3 targets the receptor-binding motif (RBM) region and exhibits broad binding to a panel of 37 RBD mutant proteins. We develop the IgG-like bispecific antibody G7-Fc using 7F3 and the cross-neutralizing antibody GW01. G7-Fc demonstrates robust neutralizing activity against all 28 tested SARS-CoV-2 variants and sarbecoviruses, providing potent prophylaxis and therapeutic efficacy against XBB.1 infection in both K18-ACE and BALB/c female mice. Cryo-EM structure analysis of the G7-Fc in complex with the Omicron XBB spike (S) trimer reveals a trimer-dimer conformation, with G7-Fc synergistically targeting two distinct RBD epitopes and blocking ACE2 binding. Comparative analysis of 7F3 and LY-CoV1404 epitopes highlights a distinct and highly conserved epitope in the RBM region bound by 7F3, facilitating neutralization of the immune-evasive Omicron variant XBB.1.16. G7-Fc holds promise as a potential prophylactic countermeasure against SARS-CoV-2, particularly against circulating and emerging variants.


Sujet(s)
Anticorps bispécifiques , Anticorps antiviraux , COVID-19 , Souris de lignée BALB C , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Animaux , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/pharmacologie , COVID-19/immunologie , COVID-19/virologie , COVID-19/prévention et contrôle , Humains , Femelle , Souris , Anticorps antiviraux/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Anticorps neutralisants/immunologie , Tests de neutralisation , Cryomicroscopie électronique , Cellules HEK293
16.
mBio ; 15(7): e0109224, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38847539

RÉSUMÉ

Herpes B virus (BV) is a zoonotic virus and belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). BV typically establishes asymptomatic infection in its natural hosts, macaque monkeys. However, in humans, BV infection causes serious neurological diseases and death. As such, BV research can only be conducted in a high containment level facility (i.e., biosafety level [BSL] 4), and the mechanisms of BV entry have not been fully elucidated. In this study, we generated a pseudotyped vesicular stomatitis virus (VSV) expressing BV glycoproteins using G-complemented VSV∆G system, which we named VSV/BVpv. We found that four BV glycoproteins (i.e., gB, gD, gH, and gL) were required for the production of a high-titer VSV/BVpv. Moreover, VSV/BVpv cell entry was dependent on the binding of gD to its cellular receptor nectin-1. Pretreatment of Vero cells with endosomal acidification inhibitors did not affect the VSV/BVpv infection. The result indicated that VSV/BVpv entry occurred by direct fusion with the plasma membrane of Vero cells and suggested that the entry pathway was similar to that of native HSV. Furthermore, we developed a VSV/BVpv-based chemiluminescence reduction neutralization test (CRNT), which detected the neutralization antibodies against BV in macaque plasma samples with high sensitivity and specificity. Crucially, the VSV/BVpv generated in this study can be used under BSL-2 condition to study the initial entry process through gD-nectin-1 interaction and the direct fusion of BV with the plasma membrane of Vero cells.IMPORTANCEHerpes B virus (BV) is a highly pathogenic zoonotic virus against humans. BV belongs to the genus Simplexvius, the same genus as human herpes simplex virus (HSV). By contrast to HSV, cell entry mechanisms of BV are not fully understood. The research procedures to manipulate infectious BV should be conducted in biosafety level (BSL)-4 facilities. As pseudotyped viruses provide a safe viral entry model because of their inability to produce infectious progeny virus, we tried to generate a pseudotyped vesicular stomatitis virus bearing BV glycoproteins (VSV/BVpv) by modification of expression constructs of BV glycoproteins, and successfully obtained VSV/BVpv with a high titer. This study has provided novel information for constructing VSV/BVpv and its usefulness to study BV infection.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Pénétration virale , Animaux , Anticorps neutralisants/immunologie , Chlorocebus aethiops , Cellules Vero , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Humains , Tests de neutralisation , Vesiculovirus/génétique , Vesiculovirus/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/métabolisme , Glycoprotéines/génétique , Glycoprotéines/immunologie , Glycoprotéines/métabolisme , Virus de la stomatite vésiculeuse de type Indiana/génétique , Virus de la stomatite vésiculeuse de type Indiana/immunologie , Protéines virales/génétique , Protéines virales/immunologie , Protéines virales/métabolisme
17.
J Virol Methods ; 328: 114959, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788979

RÉSUMÉ

In Foot-and-mouth disease (FMD) enzootic countries, periodic vaccination is the key tool in controlling the disease incidence. Active seromonitoring of the vaccinated population is critical to assess the impact of vaccination. Virus neutralization test (VNT) and enzyme-linked immunosorbent assays (ELISA) are commonly used for antibody detection. Assays like liquid phase blocking ELISA (LPBE) or solid phase competition ELISA (SPCE) are preferred as they do not require handling of live FMDV and are routinely used for seromonitoring or for vaccine potency testing; however, false positives are high in LPBE. Here we report, a monoclonal antibody (mAb) based SPCE as a potential alternate assay for antibody titration. From a panel of 12 mAbs against FMDV serotype A, two mAbs were chosen for the development of SPCE. Based on a set of 453 sera, it was demonstrated that mAb 2C4G11, mAb 6E8D11and polyclonal antibody (pAb) based SPCE had a relative sensitivity of 86.1, 86.1 and 80.3 %; and specificity of 99.6, 99.1 and 99.1 %, respectively. The correlation, repeatability, and level of agreement of the assays were high demonstrating the potential use of mAb in large scale surveillance studies and regular vaccine potency testing.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , Test ELISA , Virus de la fièvre aphteuse , Fièvre aphteuse , Sensibilité et spécificité , Sérogroupe , Test ELISA/méthodes , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Virus de la fièvre aphteuse/immunologie , Animaux , Anticorps monoclonaux/immunologie , Fièvre aphteuse/diagnostic , Fièvre aphteuse/immunologie , Fièvre aphteuse/virologie , Bovins , Tests de neutralisation/méthodes
18.
Virus Res ; 346: 199409, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38815869

RÉSUMÉ

Crimean-Congo Haemorrhagic Fever Virus (CCHFV) is spread by infected ticks or direct contact with blood, tissues and fluids from infected patients or livestock. Infection with CCHFV causes severe haemorrhagic fever in humans which is fatal in up to 83 % of cases. CCHFV is listed as a priority pathogen by the World Health Organization (WHO) and there are currently no widely-approved vaccines. Defining a serological correlate of protection against CCHFV infection would support the development of vaccines by providing a 'target threshold' for pre-clinical and clinical immunogenicity studies to achieve in subjects and potentially obviate the need for in vivo protection studies. We therefore sought to establish titratable protection against CCHFV using pooled human convalescent plasma, in a mouse model. Convalescent plasma collected from seven individuals with a known previous CCHFV virus infection were characterised using binding antibody and neutralisation assays. All plasma recognised nucleoprotein and the Gc glycoprotein, but some had a lower Gn glycoprotein response by ELISA. Pooled plasma and two individual donations from convalescent donors were administered intraperitoneally to A129 mice 24 h prior to intradermal challenge with CCHFV (strain IbAr10200). A partial protective effect was observed with all three convalescent plasmas characterised by longer survival post-challenge and reduced clinical score. These protective responses were titratable. Further characterisation of the serological reactivities within these samples will establish their value as reference materials to support assay harmonisation and accelerate vaccine development for CCHFV.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Modèles animaux de maladie humaine , Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Animaux , Fièvre hémorragique de Crimée-Congo/immunologie , Fièvre hémorragique de Crimée-Congo/prévention et contrôle , Souris , Virus de la fièvre hémorragique de Crimée-Congo/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Humains , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Femelle , Tests de neutralisation , Plasma sanguin/immunologie , Mâle
19.
Emerg Microbes Infect ; 13(1): 2359004, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38779718

RÉSUMÉ

As SARS-CoV-2 continues to spread and mutate, tracking the viral evolutionary trajectory and understanding the functional consequences of its mutations remain crucial. Here, we characterized the antibody evasion, ACE2 receptor engagement, and viral infectivity of the highly mutated SARS-CoV-2 Omicron subvariant BA.2.87.1. Compared with other Omicron subvariants, including EG.5.1 and the current predominant JN.1, BA.2.87.1 exhibits less immune evasion, reduced viral receptor engagement, and comparable infectivity in Calu-3 lung cells. Intriguingly, two large deletions (Δ15-26 and Δ136-146) in the N-terminal domain (NTD) of the spike protein facilitate subtly increased antibody evasion but significantly diminish viral infectivity. Collectively, our data support the announcement by the USA CDC that the public health risk posed by BA.2.87.1 appears to be low.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Échappement immunitaire , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Humains , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie , COVID-19/virologie , COVID-19/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Lignée cellulaire , Mutation , Tests de neutralisation
20.
Nat Commun ; 15(1): 3924, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724518

RÉSUMÉ

An effective HIV-1 vaccine must elicit broadly neutralizing antibodies (bnAbs) against highly diverse Envelope glycoproteins (Env). Since Env with the longest hypervariable (HV) loops is more resistant to the cognate bnAbs than Env with shorter HV loops, we redesigned hypervariable loops for updated Env consensus sequences of subtypes B and C and CRF01_AE. Using modeling with AlphaFold2, we reduced the length of V1, V2, and V5 HV loops while maintaining the integrity of the Env structure and glycan shield, and modified the V4 HV loop. Spacers are designed to limit strain-specific targeting. All updated Env are infectious as pseudoviruses. Preliminary structural characterization suggests that the modified HV loops have a limited impact on Env's conformation. Binding assays show improved binding to modified subtype B and CRF01_AE Env but not to subtype C Env. Neutralization assays show increases in sensitivity to bnAbs, although not always consistently across clades. Strikingly, the HV loop modification renders the resistant CRF01_AE Env sensitive to 10-1074 despite the absence of a glycan at N332.


Sujet(s)
Anticorps neutralisants , Anticorps anti-VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Produits du gène env du virus de l'immunodéficience humaine , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Humains , Produits du gène env du virus de l'immunodéficience humaine/immunologie , Produits du gène env du virus de l'immunodéficience humaine/composition chimique , Produits du gène env du virus de l'immunodéficience humaine/métabolisme , Anticorps anti-VIH/immunologie , Anticorps neutralisants/immunologie , Vaccins contre le SIDA/immunologie , Tests de neutralisation , Cellules HEK293 , Séquence consensus , Infections à VIH/virologie , Infections à VIH/immunologie , Liaison aux protéines , Épitopes/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...