Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 260
Filtrer
1.
Immunohorizons ; 8(6): 415-430, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38885041

RÉSUMÉ

The individual HLA-related susceptibility to emerging viral diseases such as COVID-19 underscores the importance of understanding how HLA polymorphism influences peptide presentation and T cell recognition. Similar to HLA-A*0101, which is one of the earliest identified HLA alleles among the human population, HLA-A*2601 possesses a similar characteristic for the binding peptide and acts as a prevalent allomorph in HLA-I. In this study, we found that, compared with HLA-A*0101, HLA-A*2601 individuals exhibit distinctive features for the T cell responses to SARS-CoV-2 and influenza virus after infection and/or vaccination. The heterogeneous T cell responses can be attributed to the distinct preference of HLA-A*2601 and HLA-A*0101 to T cell epitope motifs with negative-charged residues at the P1 and P3 positions, respectively. Furthermore, we determined the crystal structures of the HLA-A*2601 complexed to four peptides derived from SARS-CoV-2 and human papillomavirus, with one structure of HLA-A*0101 for comparison. The shallow pocket C of HLA-A*2601 results in the promiscuous presentation of peptides with "switchable" bulged conformations because of the secondary anchor in the median portion. Notably, the hydrogen bond network formed between the negative-charged P1 anchors and the HLA-A*2601-specific residues lead to a "closed" conformation and solid placement for the P1 secondary anchor accommodation in pocket A. This insight sheds light on the intricate relationship between HLA I allelic allomorphs, peptide binding, and the immune response and provides valuable implications for understanding disease susceptibility and potential vaccine design.


Sujet(s)
COVID-19 , Déterminants antigéniques des lymphocytes T , SARS-CoV-2 , Humains , Déterminants antigéniques des lymphocytes T/immunologie , Déterminants antigéniques des lymphocytes T/génétique , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , COVID-19/immunologie , COVID-19/virologie , Antigènes HLA-A/immunologie , Antigènes HLA-A/génétique , Antigènes HLA-A/métabolisme , Antigènes HLA-A/composition chimique , Peptides/immunologie , Peptides/composition chimique , Allèles , Antigène HLA-A1
2.
Proc Natl Acad Sci U S A ; 121(24): e2400163121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38830098

RÉSUMÉ

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease with a high fatality rate of up to 30% caused by SFTS virus (SFTSV). However, no specific vaccine or antiviral therapy has been approved for clinical use. To develop an effective treatment, we isolated a panel of human monoclonal antibodies (mAbs). SF5 and SF83 are two neutralizing mAbs that recognize two viral glycoproteins (Gn and Gc), respectively. We found that their epitopes are closely located, and we then engineered them as several bispecific antibodies (bsAbs). Neutralization and animal experiments indicated that bsAbs display more potent protective effects than the parental mAbs, and the cryoelectron microscopy structure of a bsAb3 Fab-Gn-Gc complex elucidated the mechanism of protection. In vivo virus passage in the presence of antibodies indicated that two bsAbs resulted in less selective pressure and could efficiently bind to all single parental mAb-escape mutants. Furthermore, epitope analysis of the protective mAbs against SFTSV and RVFV indicated that they are all located on the Gn subdomain I, where may be the hot spots in the phleboviruses. Collectively, these data provide potential therapeutic agents and molecular basis for the rational design of vaccines against SFTSV infection.


Sujet(s)
Anticorps bispécifiques , Anticorps neutralisants , Anticorps antiviraux , Phlebovirus , Animaux , Anticorps bispécifiques/immunologie , Souris , Anticorps neutralisants/immunologie , Phlebovirus/immunologie , Humains , Anticorps antiviraux/immunologie , Glycoprotéines/immunologie , Anticorps monoclonaux/immunologie , Épitopes/immunologie , Modèles animaux de maladie humaine , Syndrome de fièvre sévère avec thrombocytopénie/immunologie , Syndrome de fièvre sévère avec thrombocytopénie/prévention et contrôle
3.
Cell Rep ; 43(6): 114338, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38850530

RÉSUMÉ

The game between therapeutic monoclonal antibodies (mAbs) and continuously emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants has favored the virus, as most therapeutic mAbs have been evaded. Addressing this challenge, we systematically explored a reproducible bispecific antibody (bsAb)-dependent synergistic effect in this study. It could effectively restore the neutralizing activity of the bsAb when any of its single mAbs is escaped by variants. This synergy is primarily attributed to the binding angle of receptor-binding domain (RBD)-5, facilitating inter-spike cross-linking and promoting cryptic epitope exposure that classical antibody cocktails cannot achieve. Furthermore, RBD-5 with RBD-2, RBD-6, and RBD-7, alongside RBD-8, also exhibit significantly enhanced effects. This study not only shifts the paradigm in understanding antibody interactions but paves the way for developing more effective therapeutic antibodies against rapidly mutating SARS-CoV-2, with Dia-19 already showing promise against emerging variants like BA.2.86, EG.5.1, and JN.1.


Sujet(s)
Anticorps bispécifiques , Anticorps neutralisants , Anticorps antiviraux , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , SARS-CoV-2/immunologie , Humains , Anticorps bispécifiques/immunologie , Anticorps bispécifiques/pharmacologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/virologie , COVID-19/thérapie , Glycoprotéine de spicule des coronavirus/immunologie , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie , Épitopes/immunologie , Liaison aux protéines , Animaux
4.
Pharmacol Res ; 204: 107217, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38777110

RÉSUMÉ

The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway functions as a central hub for transmitting signals from more than 50 cytokines, playing a pivotal role in maintaining hematopoiesis, immune balance, and tissue homeostasis. Dysregulation of this pathway has been implicated in various diseases, including immunodeficiency, autoimmune conditions, hematological disorders, and certain cancers. Proteins within this pathway have emerged as effective therapeutic targets for managing these conditions, with various approaches developed to modulate key nodes in the signaling process, spanning from receptor engagement to transcription factor activation. Following the success of JAK inhibitors such as tofacitinib for RA treatment and ruxolitinib for managing primary myelofibrosis, the pharmaceutical industry has obtained approvals for over 10 small molecule drugs targeting the JAK-STAT pathway and many more are at various stages of clinical trials. In this review, we consolidate key strategies employed in drug discovery efforts targeting this pathway, with the aim of contributing to the collective understanding of small molecule interventions in the context of JAK-STAT signaling. We aspire that our endeavors will contribute to advancing the development of innovative and efficacious treatments for a range of diseases linked to this pathway dysregulation.


Sujet(s)
Découverte de médicament , Janus kinases , Facteurs de transcription STAT , Transduction du signal , Humains , Janus kinases/métabolisme , Janus kinases/antagonistes et inhibiteurs , Facteurs de transcription STAT/métabolisme , Facteurs de transcription STAT/antagonistes et inhibiteurs , Découverte de médicament/méthodes , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/pharmacologie , Thérapie moléculaire ciblée
5.
Signal Transduct Target Ther ; 9(1): 131, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740785

RÉSUMÉ

Almost all the neutralizing antibodies targeting the receptor-binding domain (RBD) of spike (S) protein show weakened or lost efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged or emerging variants, such as Omicron and its sub-variants. This suggests that highly conserved epitopes are crucial for the development of neutralizing antibodies. Here, we present one nanobody, N235, displaying broad neutralization against the SARS-CoV-2 prototype and multiple variants, including the newly emerged Omicron and its sub-variants. Cryo-electron microscopy demonstrates N235 binds a novel, conserved, cryptic epitope in the N-terminal domain (NTD) of the S protein, which interferes with the RBD in the neighboring S protein. The neutralization mechanism interpreted via flow cytometry and Western blot shows that N235 appears to induce the S1 subunit shedding from the trimeric S complex. Furthermore, a nano-IgM construct (MN235), engineered by fusing N235 with the human IgM Fc region, displays prevention via inducing S1 shedding and cross-linking virus particles. Compared to N235, MN235 exhibits varied enhancement in neutralization against pseudotyped and authentic viruses in vitro. The intranasal administration of MN235 in low doses can effectively prevent the infection of Omicron sub-variant BA.1 and XBB in vivo, suggesting that it can be developed as a promising prophylactic antibody to cope with the ongoing and future infection.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , COVID-19 , Épitopes , Immunoglobuline M , SARS-CoV-2 , Anticorps à domaine unique , Glycoprotéine de spicule des coronavirus , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Humains , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/génétique , Anticorps à domaine unique/composition chimique , Anticorps à domaine unique/pharmacologie , Épitopes/immunologie , Épitopes/génétique , Épitopes/composition chimique , Animaux , COVID-19/immunologie , COVID-19/virologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/composition chimique , Anticorps neutralisants/immunologie , Anticorps neutralisants/composition chimique , Anticorps neutralisants/génétique , Immunoglobuline M/immunologie , Immunoglobuline M/génétique , Souris , Domaines protéiques , Cryomicroscopie électronique
6.
Structure ; 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38626767

RÉSUMÉ

Nectin and nectin-like (Necl) co-receptor axis, comprised of receptors DNAM-1, TIGIT, CD96, PVRIG, and nectin/Necl ligands, is gaining prominence in immuno-oncology. Within this axis, the inhibitory receptor PVRIG recognizes Nectin-2 with high affinity, but the underlying molecular basis remains unknown. By determining the crystal structure of PVRIG in complex with Nectin-2, we identified a unique CC' loop in PVRIG, which complements the double-lock-and-key binding mode and contributes to its high affinity for Nectin-2. The association of the corresponding charged residues in the F-strands explains the ligand selectivity of PVRIG toward Nectin-2 but not for Necl-5. Moreover, comprehensive comparisons of the binding capacities between co-receptors and ligands provide innovative insights into the intra-axis immunoregulatory mechanism. Taken together, these findings broaden our understanding of immune recognition and regulation mediated by nectin/Necl co-receptors and provide a rationale for the development of immunotherapeutic strategies targeting the nectin/Necl axis.

7.
EMBO J ; 43(8): 1484-1498, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38467833

RÉSUMÉ

Since SARS-CoV-2 Omicron variant emerged, it is constantly evolving into multiple sub-variants, including BF.7, BQ.1, BQ.1.1, XBB, XBB.1.5 and the recently emerged BA.2.86 and JN.1. Receptor binding and immune evasion are recognized as two major drivers for evolution of the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein. However, the underlying mechanism of interplay between two factors remains incompletely understood. Herein, we determined the structures of human ACE2 complexed with BF.7, BQ.1, BQ.1.1, XBB and XBB.1.5 RBDs. Based on the ACE2/RBD structures of these sub-variants and a comparison with the known complex structures, we found that R346T substitution in the RBD enhanced ACE2 binding upon an interaction with the residue R493, but not Q493, via a mechanism involving long-range conformation changes. Furthermore, we found that R493Q and F486V exert a balanced impact, through which immune evasion capability was somewhat compromised to achieve an optimal receptor binding. We propose a "two-steps-forward and one-step-backward" model to describe such a compromise between receptor binding affinity and immune evasion during RBD evolution of Omicron sub-variants.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/génétique , Angiotensin-converting enzyme 2 , Glycoprotéine de spicule des coronavirus/génétique , Anticorps
8.
Nat Immunol ; 25(2): 307-315, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38182667

RÉSUMÉ

The global outbreak of the mpox virus (MPXV) in 2022 highlights the urgent need for safer and more accessible new-generation vaccines. Here, we used a structure-guided multi-antigen fusion strategy to design a 'two-in-one' immunogen based on the single-chain dimeric MPXV extracellular enveloped virus antigen A35 bivalently fused with the intracellular mature virus antigen M1, called DAM. DAM preserved the natural epitope configuration of both components and showed stronger A35-specific and M1-specific antibody responses and in vivo protective efficacy against vaccinia virus (VACV) compared to co-immunization strategies. The MPXV-specific neutralizing antibodies elicited by DAM were 28 times higher than those induced by live VACV vaccine. Aluminum-adjuvanted DAM vaccines protected mice from a lethal VACV challenge with a safety profile, and pilot-scale production confirmed the high yield and purity of DAM. Thus, our study provides innovative insights and an immunogen candidate for the development of alternative vaccines against MPXV and other orthopoxviruses.


Sujet(s)
Virus de la variole simienne , Vaccins , Animaux , Souris , Protéines de l'enveloppe virale , Anticorps antiviraux , Virus de la vaccine , Antigènes viraux , Immunité
11.
Nat Commun ; 14(1): 6389, 2023 10 12.
Article de Anglais | MEDLINE | ID: mdl-37828002

RÉSUMÉ

KRAS mutations are broadly recognized as promising targets for tumor therapy. T cell receptors (TCRs) can specifically recognize KRAS mutant neoantigens presented by human lymphocyte antigen (HLA) and mediate T cell responses to eliminate tumor cells. In the present study, we identify two TCRs specific for the 9-mer KRAS-G12V mutant neoantigen in the context of HLA-A*11:01. The TCR-T cells are constructed and display cytokine secretion and cytotoxicity upon co-culturing with varied tumor cells expressing the KRAS-G12V mutation. Moreover, 1-2C TCR-T cells show anti-tumor activity in preclinical models in female mice. The 9-mer KRAS-G12V mutant peptide exhibits a distinct conformation from the 9-mer wildtype peptide and its 10-mer counterparts. Specific recognition of the G12V mutant by TCR depends both on distinct conformation from wildtype peptide and on direct interaction with residues from TCRs. Our study reveals the mechanisms of presentation and TCR recognition of KRAS-G12V mutant peptide and describes TCRs with therapeutic potency for tumor immunotherapy.


Sujet(s)
Tumeurs , Protéines proto-oncogènes p21(ras) , Femelle , Humains , Animaux , Souris , Protéines proto-oncogènes p21(ras)/génétique , Antigènes néoplasiques , Récepteurs aux antigènes des cellules T/métabolisme , Peptides/composition chimique , Thérapie cellulaire et tissulaire
12.
Nat Commun ; 14(1): 6333, 2023 10 10.
Article de Anglais | MEDLINE | ID: mdl-37816705

RÉSUMÉ

The severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne human-infecting bunyavirus, which utilizes two envelope glycoproteins, Gn and Gc, to enter host cells. However, the structure and organization of these glycoproteins on virion surface are not yet known. Here we describe the structure of SFTSV determined by single particle reconstruction, which allows mechanistic insights into bunyavirus assembly at near-atomic resolution. The SFTSV Gn and Gc proteins exist as heterodimers and further assemble into pentameric and hexameric peplomers, shielding the Gc fusion loops by both intra- and inter-heterodimer interactions. Individual peplomers are associated mainly through the ectodomains, in which the highly conserved glycans on N914 of Gc play a crucial role. This elaborate assembly stabilizes Gc in the metastable prefusion conformation and creates some cryptic epitopes that are only accessible in the intermediate states during virus entry. These findings provide an important basis for developing vaccines and therapeutic drugs.


Sujet(s)
Orthobunyavirus , Phlebovirus , Syndrome de fièvre sévère avec thrombocytopénie , Humains , Protéines de l'enveloppe virale/métabolisme , Cryomicroscopie électronique , Glycoprotéines/métabolisme
13.
Zool Res ; 44(6): 1015-1025, 2023 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-37804113

RÉSUMÉ

Following the outbreak of coronavirus disease 2019 (COVID-19), several severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-related coronaviruses have been discovered. Previous research has identified a novel lineage of SARS-CoV-2-related CoVs in bats, including RsYN04, which recognizes human angiotensin-converting enzyme 2 (ACE2) and thus poses a potential threat to humans. Here, we screened the binding of the RsYN04 receptor-binding domain (RBD) to ACE2 orthologs from 52 animal species and found that the virus showed a narrower ACE2-binding spectrum than SARS-CoV-2. However, the presence of the T484W mutation in the RsYN04 RBD broadened its range. We also evaluated 44 SARS-CoV-2 antibodies targeting seven epitope communities in the SARS-CoV-2 RBD, together with serum obtained from COVID-19 convalescents and vaccinees, to determine their cross-reaction against RsYN04. Results showed that no antibodies, except for the RBD-6 and RBD-7 classes, bound to the RsYN04 RBD, indicating substantial immune differences from SARS-CoV-2. Furthermore, the structure of the RsYN04 RBD in complex with cross-reactive antibody S43 in RBD-7 revealed a potently broad epitope for the development of therapeutics and vaccines. Our findings suggest RsYN04 and other viruses belonging to the same clade have the potential to infect several species, including humans, highlighting the necessity for viral surveillance and development of broad anti-coronavirus countermeasures.


Sujet(s)
COVID-19 , Chiroptera , Animaux , Humains , SARS-CoV-2 , COVID-19/médecine vétérinaire , Angiotensin-converting enzyme 2 , Chiroptera/génétique , Anticorps antiviraux , Épitopes
14.
Cell ; 186(19): 4074-4084.e11, 2023 09 14.
Article de Anglais | MEDLINE | ID: mdl-37669665

RÉSUMÉ

H3N8 avian influenza viruses (AIVs) in China caused two confirmed human infections in 2022, followed by a fatal case reported in 2023. H3N8 viruses are widespread in chicken flocks; however, the zoonotic features of H3N8 viruses are poorly understood. Here, we demonstrate that H3N8 viruses were able to infect and replicate efficiently in organotypic normal human bronchial epithelial (NHBE) cells and lung epithelial (Calu-3) cells. Human isolates of H3N8 virus were more virulent and caused severe pathology in mice and ferrets, relative to chicken isolates. Importantly, H3N8 virus isolated from a patient with severe pneumonia was transmissible between ferrets through respiratory droplets; it had acquired human-receptor-binding preference and amino acid substitution PB2-E627K necessary for airborne transmission. Human populations, even when vaccinated against human H3N2 virus, appear immunologically naive to emerging mammalian-adapted H3N8 AIVs and could be vulnerable to infection at epidemic or pandemic proportion.


Sujet(s)
Sous-type H3N8 du virus de la grippe A , Grippe humaine , Animaux , Humains , Souris , Poulets , Furets , Sous-type H3N2 du virus de la grippe A , Gouttelettes et aérosols respiratoires
15.
J Virol ; 97(9): e0050523, 2023 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-37676003

RÉSUMÉ

SARS-CoV-2 has been expanding its host range, among which the white-tailed deer (WTD), Odocoileus virginianus, became the first wildlife species infected on a large scale and might serve as a host reservoir for variants of concern (VOCs) in case no longer circulating in humans. In this study, we comprehensively assessed the binding of the WTD angiotensin-converting enzyme 2 (ACE2) receptor to the spike (S) receptor-binding domains (RBDs) from the SARS-CoV-2 prototype (PT) strain and multiple variants. We found that WTD ACE2 could be broadly recognized by all of the tested RBDs. We further determined the complex structures of WTD ACE2 with PT, Omicron BA.1, and BA.4/5 S trimer. Detailed structural comparison revealed the important roles of RBD residues on 486, 498, and 501 sites for WTD ACE2 binding. This study deepens our understanding of the interspecies transmission mechanisms of SARS-CoV-2 and further addresses the importance of constant monitoring on SARS-CoV-2 infections in wild animals. IMPORTANCE Even if we manage to eliminate the virus among humans, it will still circulate among wildlife and continuously be transmitted back to humans. A recent study indicated that WTD may serve as reservoir for nearly extinct SARS-CoV-2 strains. Therefore, it is critical to evaluate the binding abilities of SARS-CoV-2 variants to the WTD ACE2 receptor and elucidate the molecular mechanisms of binding of the RBDs to assess the risk of spillback events.

16.
Nature ; 622(7983): 603-610, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37699521

RÉSUMÉ

Non-segmented negative-strand RNA viruses, including Ebola virus (EBOV), rabies virus, human respiratory syncytial virus and pneumoviruses, can cause respiratory infections, haemorrhagic fever and encephalitis in humans and animals, and are considered a substantial health and economic burden worldwide1. Replication and transcription of the viral genome are executed by the large (L) polymerase, which is a promising target for the development of antiviral drugs. Here, using the L polymerase of EBOV as a representative, we show that de novo replication of L polymerase is controlled by the specific 3' leader sequence of the EBOV genome in an enzymatic assay, and that formation of at least three base pairs can effectively drive the elongation process of RNA synthesis independent of the specific RNA sequence. We present the high-resolution structures of the EBOV L-VP35-RNA complex and show that the 3' leader RNA binds in the template entry channel with a distinctive stable bend conformation. Using mutagenesis assays, we confirm that the bend conformation of the RNA is required for the de novo replication activity and reveal the key residues of the L protein that stabilize the RNA conformation. These findings provide a new mechanistic understanding of RNA synthesis for polymerases of non-segmented negative-strand RNA viruses, and reveal important targets for the development of antiviral drugs.


Sujet(s)
Ebolavirus , ARN viral , RNA replicase , Réplication virale , Animaux , Humains , Antiviraux/pharmacologie , Ebolavirus/enzymologie , Ebolavirus/génétique , Ebolavirus/croissance et développement , Fièvre hémorragique à virus Ebola/virologie , ARN viral/biosynthèse , ARN viral/composition chimique , ARN viral/génétique , ARN viral/métabolisme , RNA replicase/antagonistes et inhibiteurs , RNA replicase/métabolisme , Génome viral , Conformation d'acide nucléique , Mutagenèse , Stabilité de l'ARN
17.
PLoS Pathog ; 19(9): e1011659, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37721934

RÉSUMÉ

SARS-CoV-2 variants with severe immune evasion are a major challenge for COVID-19 prevention, especially the circulating Omicron XBB/BQ.1.1/BF.7 strains. Thus, the next-generation of broad-spectrum vaccines are urgently needed. Previously, we developed a COVID-19 protein subunit vaccine, ZF2001, based on the RBD-homodimer as the immunogen. To adapt SARS-CoV-2 variants, we developed chimeric RBD-heterodimers to induce broad immune responses. In this study, we further explored the concept of tandem RBD homotrimer and heterotrimer. Prototype SARS-CoV-2 RBD-homotrimer, prototype-Delta-BA.1 (PDO) RBD-heterotrimer and Delta-BA.2-BA.5 (DBA2BA5) RBD-heterotrimer were designed. Biochemical and cryo-EM structural characterization demonstrated total epitope exposure of the RBD-trimers. In mouse experiments, PDO and DBA2BA5 elicited broad SARS-CoV-2 neutralization. Potent protection against SARS-CoV-2 variants was observed in challenge assays and was correlated with neutralizing antibody titer. This study validated the design strategy of tandem RBD-heterotrimers as multivalent immunogens and presented a promising vaccine candidate, DBA2BA5, eliciting broad-spectrum immune responses, including against the circulating XBB/BF.7/BQ.1.1.


Sujet(s)
COVID-19 , Vaccins , Animaux , Souris , SARS-CoV-2/génétique , COVID-19/prévention et contrôle , Anticorps neutralisants , Anticorps antiviraux
18.
J Med Virol ; 95(7): e28954, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37493180

RÉSUMÉ

At present, the horse or human rabies immunoglobulin (RIG) used for postexposure prevention of human rabies (PEP) has high cost and limited availability. It is strongly encouraged to replace RIG with equivalent or more effective and safer products. Mouse and human monoclonal antibodies have been shown to protect rodents from lethal rabies virus (RABV) attacks. In this study, we reported a human-mouse chimeric monoclonal antibody, 12-2A12, which showed a strong neutralization potency and a wide breadth against multiple street viruses of RABV in vitro. The antibody binded the viral glycoprotein (G) with nanomolar affinity. The complex structure of 12-2A12 bound to RABV G revealed that the antibody recognizes an epitope that partially overlaps with the recognition region for the nicotinic acetylcholine receptor (nAChR). The antibody therefore would interfere with the nAChR/G interaction to block the viral receptor binding. In addition, comparison of our complex structure with the G structure in the acidic state reveals a clear steric clash, highlighting that the antibody would further prevent the conformational changes of the viral glycoprotein that are essential for membrane fusion. In light of these functional and structural data, we believe that 12-2A12 might be developed to be included in an antibody cocktail for potential use in human rabies PEP.


Sujet(s)
Virus de la rage , Rage (maladie) , Humains , Animaux , Souris , Equus caballus , Rage (maladie)/prévention et contrôle , Anticorps antiviraux , Glycoprotéines , Anticorps monoclonaux , Facteurs immunologiques/métabolisme , Immunosuppresseurs
19.
Nat Commun ; 14(1): 4405, 2023 07 21.
Article de Anglais | MEDLINE | ID: mdl-37479708

RÉSUMÉ

Multiple SARS-CoV-2 Omicron sub-variants, such as BA.2, BA.2.12.1, BA.4, and BA.5, emerge one after another. BA.5 has become the dominant strain worldwide. Additionally, BA.2.75 is significantly increasing in some countries. Exploring their receptor binding and interspecies transmission risk is urgently needed. Herein, we examine the binding capacities of human and other 28 animal ACE2 orthologs covering nine orders towards S proteins of these sub-variants. The binding affinities between hACE2 and these sub-variants remain in the range as that of previous variants of concerns (VOCs) or interests (VOIs). Notably, R493Q reverse mutation enhances the bindings towards ACE2s from humans and many animals closely related to human life, suggesting an increased risk of cross-species transmission. Structures of S/hACE2 or RBD/hACE2 complexes for these sub-variants and BA.2 S binding to ACE2 of mouse, rat or golden hamster are determined to reveal the molecular basis for receptor binding and broader interspecies recognition.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Cricetinae , Humains , Animaux , Souris , Rats , SARS-CoV-2/génétique , Mesocricetus , Mutation
20.
Front Immunol ; 14: 1160283, 2023.
Article de Anglais | MEDLINE | ID: mdl-37234155

RÉSUMÉ

Introduction: The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has been posing a severe threat to global public health. Although broadly neutralizing antibodies have been used to prevent or treat corona virus disease 2019 (COVID-19), new emerging variants have been proven resistant to these antibodies. Methods: In this study, we isolated receptor binding domain (RBD)-specific memory B cells using single-cell sorting method from two COVID-19 convalescents and expressed the antibody to test their neutralizing activity against diverse SARS-CoV-2 variants. Then, we resolved antibody-RBD complex structures of potent RBD-specific neutralizing antibodies by X-ray diffraction method. Finally, we analyzed the whole antibody repertoires of the two donors and studied the evolutionary pathway of potent neutralizing antibodies. Results and discussion: We identified three potent RBD-specific neutralizing antibodies (1D7, 3G10 and 3C11) from two COVID-19 convalescents that neutralized authentic SARS-CoV-2 WH-1 and Delta variant, and one of them, 1D7, presented broadly neutralizing activity against WH-1, Beta, Gamma, Delta and Omicron authentic viruses. The resolved antibody-RBD complex structures of two antibodies, 3G10 and 3C11, indicate that both of them interact with the external subdomain of the RBD and that they belong to the RBD-1 and RBD-4 communities, respectively. From the antibody repertoire analysis, we found that the CDR3 frequencies of the light chain, which shared high degrees of amino acid identity with these three antibodies, were higher than those of the heavy chain. This research will contribute to the development of RBD-specific antibody-based drugs and immunogens against multiple variants.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , Anticorps neutralisants à large spectre , Anticorps neutralisants
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...