Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 133
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(22): e2310677121, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38753503

RÉSUMÉ

Seasonal and pandemic-associated influenza strains cause highly contagious viral respiratory infections that can lead to severe illness and excess mortality. Here, we report on the optimization of our small-molecule inhibitor F0045(S) targeting the influenza hemagglutinin (HA) stem with our Sulfur-Fluoride Exchange (SuFEx) click chemistry-based high-throughput medicinal chemistry (HTMC) strategy. A combination of SuFEx- and amide-based lead molecule diversification and structure-guided design led to identification and validation of ultrapotent influenza fusion inhibitors with subnanomolar EC50 cellular antiviral activity against several influenza A group 1 strains. X-ray structures of six of these compounds with HA indicate that the appended moieties occupy additional pockets on the HA surface and increase the binding interaction, where the accumulation of several polar interactions also contributes to the improved affinity. The compounds here represent the most potent HA small-molecule inhibitors to date. Our divergent HTMC platform is therefore a powerful, rapid, and cost-effective approach to develop bioactive chemical probes and drug-like candidates against viral targets.


Sujet(s)
Antiviraux , Glycoprotéine hémagglutinine du virus influenza , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Humains , Antiviraux/pharmacologie , Antiviraux/composition chimique , Chimie pharmaceutique/méthodes , Tests de criblage à haut débit/méthodes , Grippe humaine/traitement médicamenteux , Grippe humaine/virologie , Cristallographie aux rayons X/méthodes , Chimie click/méthodes , Animaux , Virus de la grippe A/effets des médicaments et des substances chimiques , Cellules rénales canines Madin-Darby , Inhibiteurs des protéines de fusion virale/pharmacologie , Inhibiteurs des protéines de fusion virale/composition chimique , Chiens
2.
Structure ; 32(6): 766-779.e7, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38537643

RÉSUMÉ

Endolysosomal exonucleases PLD3 and PLD4 (phospholipases D3 and D4) are associated with autoinflammatory and autoimmune diseases. We report structures of these enzymes, and the molecular basis of their catalysis. The structures reveal an intra-chain dimer topology forming a basic active site at the interface. Like other PLD superfamily members, PLD3 and PLD4 carry HxKxxxxD/E motifs and participate in phosphodiester-bond cleavage. The enzymes digest ssDNA and ssRNA in a 5'-to-3' manner and are blocked by 5'-phosphorylation. We captured structures in apo, intermediate, and product states and revealed a "link-and-release" two-step catalysis. We also unexpectedly demonstrated phosphatase activity via a covalent 3-phosphohistidine intermediate. PLD4 contains an extra hydrophobic clamp that stabilizes substrate and could affect oligonucleotide substrate preference and product release. Biochemical and structural analysis of disease-associated mutants of PLD3/4 demonstrated reduced enzyme activity or thermostability and the possible basis for disease association. Furthermore, these findings provide insight into therapeutic design.


Sujet(s)
Domaine catalytique , Modèles moléculaires , Phospholipase D , Phospholipase D/métabolisme , Phospholipase D/composition chimique , Phospholipase D/génétique , Humains , Spécificité du substrat , Cristallographie aux rayons X , Mutation , Lysosomes/métabolisme , Lysosomes/enzymologie , Phosphorylation , ADN simple brin/métabolisme , ADN simple brin/composition chimique , Multimérisation de protéines , Liaison aux protéines , Exodeoxyribonucleases
3.
Cell Host Microbe ; 32(2): 261-275.e4, 2024 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-38307019

RÉSUMÉ

Hemagglutinins (HAs) from human influenza viruses descend from avian progenitors that bind α2-3-linked sialosides and must adapt to glycans with α2-6-linked sialic acids on human airway cells to transmit within the human population. Since their introduction during the 1968 pandemic, H3N2 viruses have evolved over the past five decades to preferentially recognize human α2-6-sialoside receptors that are elongated through addition of poly-LacNAc. We show that more recent H3N2 viruses now make increasingly complex interactions with elongated receptors while continuously selecting for strains maintaining this phenotype. This change in receptor engagement is accompanied by an extension of the traditional receptor-binding site to include residues in key antigenic sites on the surface of HA trimers. These results help explain the propensity for selection of antigenic variants, leading to vaccine mismatching, when H3N2 viruses are propagated in chicken eggs or cells that do not contain such receptors.


Sujet(s)
Sous-type H3N2 du virus de la grippe A , Grippe humaine , Animaux , Humains , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/métabolisme , Récepteurs viraux/composition chimique , Acides sialiques/métabolisme , Polyosides/métabolisme , Poulets , Glycoprotéine hémagglutinine du virus influenza
4.
bioRxiv ; 2023 Nov 20.
Article de Anglais | MEDLINE | ID: mdl-38045427

RÉSUMÉ

Endolysosomal exonucleases PLD3 and PLD4 (phospholipases D3 and D4) are associated with autoinflammatory and autoimmune diseases. We report structures of these enzymes, and the molecular basis of their catalysis. The structures reveal an intra-chain dimer topology forming a basic active site at the interface. Like other PLD superfamily members, PLD3 and PLD4 carry HxKxxxxD/E motifs and participate in phosphodiester-bond cleavage. The enzymes digest ssDNA and ssRNA in a 5'-to-3' manner and are blocked by 5'-phosphorylation. We captured structures in apo, intermediate, and product states and revealed a 'link-and-release' two-step catalysis. We also unexpectedly demonstrated phosphatase activity via a covalent 3' phosphistidine intermediate. PLD4 contains an extra hydrophobic clamp that stabilizes substrate and could affect oligonucleotide substrate preference and product release. Biochemical and structural analysis of disease-associated mutants of PLD3/4 demonstrated reduced enzyme activity or thermostability and the possible basis for disease association. Furthermore, these findings provide insight into therapeutic design.

5.
Sci Adv ; 9(37): eadi4753, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37703367

RÉSUMÉ

Seasonal influenza virus vaccines are effective when they are well matched to circulating strains. Because of antigenic drift/change in the immunodominant hemagglutinin (HA) head domain, annual vaccine reformulations are necessary to maintain a match with circulating strains. In addition, seasonal vaccines provide little to no protection against newly emerging pandemic strains. Sequential vaccination with chimeric HA (cHA) constructs has been proven to direct the immune response toward the immunosubdominant but more conserved HA stalk domain. In this study, we show that immunization with group 2 cHA split vaccines in combination with the CpG 1018 adjuvant elicits broadly cross-reactive antibodies against all group 2 HAs, as well as systemic and local antigen-specific T cell responses. Antibodies elicited after sequential vaccination are directed to conserved regions of the HA such as the stalk and the trimer interface and also to the N2 neuraminidase (NA). Immunized mice were fully protected from challenge with a broad panel of influenza A viruses.


Sujet(s)
Virus de la grippe A , Vaccins antigrippaux , Animaux , Souris , Hémagglutinines , Anticorps , Vaccination , Épitopes immunodominants
6.
Sci Signal ; 16(798): eabk3516, 2023 08 15.
Article de Anglais | MEDLINE | ID: mdl-37582161

RÉSUMÉ

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) underscores the need for strategies to rapidly develop neutralizing monoclonal antibodies that can function as prophylactic and therapeutic agents and to help guide vaccine design. Here, we demonstrate that engineering approaches can be used to refocus an existing antibody that neutralizes one virus but not a related virus. Through a rapid affinity maturation strategy, we engineered CR3022, a SARS-CoV-1-neutralizing antibody, to bind to the receptor binding domain of SARS-CoV-2 with >1000-fold increased affinity. The engineered CR3022 neutralized SARS-CoV-2 and provided prophylactic protection from viral challenge in a small animal model of SARS-CoV-2 infection. Deep sequencing throughout the engineering process paired with crystallographic analysis of engineered CR3022 elucidated the molecular mechanisms by which the antibody can accommodate sequence differences in the epitopes between SARS-CoV-1 and SARS-CoV-2. This workflow provides a blueprint for the rapid broadening of neutralization of an antibody from one virus to closely related but resistant viruses.


Sujet(s)
COVID-19 , SARS-CoV-2 , Animaux , SARS-CoV-2/génétique , COVID-19/prévention et contrôle , Anticorps antiviraux , Tests de neutralisation , Anticorps neutralisants
7.
Immunity ; 56(8): 1927-1938.e8, 2023 08 08.
Article de Anglais | MEDLINE | ID: mdl-37506693

RÉSUMÉ

Neuraminidase (NA) is one of the two influenza virus surface glycoproteins, and antibodies that target it are an independent correlate of protection. However, our current understanding of NA antigenicity is incomplete. Here, we describe human monoclonal antibodies (mAbs) from a patient with a pandemic H1N1 virus infection in 2009. Two mAbs exhibited broad reactivity and inhibited NA enzyme activity of seasonal H1N1 viruses circulating before and after 2009, as well as viruses with avian or swine N1s. The mAbs provided robust protection from lethal challenge with human H1N1 and avian H5N1 viruses in mice, and both target an epitope on the lateral face of NA. In summary, we identified two broadly protective NA antibodies that share a novel epitope, inhibited NA activity, and provide protection against virus challenge in mice. Our work reaffirms that NA should be included as a target in future broadly protective or universal influenza virus vaccines.


Sujet(s)
Anticorps monoclonaux , Anticorps antiviraux , Sous-type H1N1 du virus de la grippe A , Grippe humaine , Sialidase , Anticorps monoclonaux/isolement et purification , Anticorps monoclonaux/métabolisme , Anticorps antiviraux/isolement et purification , Anticorps antiviraux/métabolisme , Sialidase/composition chimique , Sialidase/métabolisme , Humains , Fragments Fab d'immunoglobuline/composition chimique , Cryomicroscopie électronique , Épitopes , Souris de lignée BALB C , Animaux , Souris , Grippe humaine/traitement médicamenteux , Modèles animaux de maladie humaine
8.
Proc Natl Acad Sci U S A ; 120(24): e2216612120, 2023 06 13.
Article de Anglais | MEDLINE | ID: mdl-37276407

RÉSUMÉ

Nanobodies bind a target antigen with a kinetic profile similar to a conventional antibody, but exist as a single heavy chain domain that can be readily multimerized to engage antigen via multiple interactions. Presently, most nanobodies are produced by immunizing camelids; however, platforms for animal-free production are growing in popularity. Here, we describe the development of a fully synthetic nanobody library based on an engineered human VH3-23 variable gene and a multispecific antibody-like format designed for biparatopic target engagement. To validate our library, we selected nanobodies against the SARS-CoV-2 receptor-binding domain and employed an on-yeast epitope binning strategy to rapidly map the specificities of the selected nanobodies. We then generated antibody-like molecules by replacing the VH and VL domains of a conventional antibody with two different nanobodies, designed as a molecular clamp to engage the receptor-binding domain biparatopically. The resulting bispecific tetra-nanobody immunoglobulins neutralized diverse SARS-CoV-2 variants with potencies similar to antibodies isolated from convalescent donors. Subsequent biochemical analyses confirmed the accuracy of the on-yeast epitope binning and structures of both individual nanobodies, and a tetra-nanobody immunoglobulin revealed that the intended mode of interaction had been achieved. This overall workflow is applicable to nearly any protein target and provides a blueprint for a modular workflow for the development of multispecific molecules.


Sujet(s)
COVID-19 , Anticorps à domaine unique , Humains , Anticorps à domaine unique/composition chimique , Saccharomyces cerevisiae/métabolisme , SARS-CoV-2 , Anticorps , Épitopes
10.
bioRxiv ; 2023 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-37162858

RÉSUMÉ

Developing broad coronavirus vaccines requires identifying and understanding the molecular basis of broadly neutralizing antibody (bnAb) spike sites. In our previous work, we identified sarbecovirus spike RBD group 1 and 2 bnAbs. We have now shown that many of these bnAbs can still neutralize highly mutated SARS-CoV-2 variants, including the XBB.1.5. Structural studies revealed that group 1 bnAbs use recurrent germline-encoded CDRH3 features to interact with a conserved RBD region that overlaps with class 4 bnAb site. Group 2 bnAbs recognize a less well-characterized "site V" on the RBD and destabilize spike trimer. The site V has remained largely unchanged in SARS-CoV-2 variants and is highly conserved across diverse sarbecoviruses, making it a promising target for broad coronavirus vaccine development. Our findings suggest that targeted vaccine strategies may be needed to induce effective B cell responses to escape resistant subdominant spike RBD bnAb sites.

11.
Sci Transl Med ; 15(695): eadg7404, 2023 05 10.
Article de Anglais | MEDLINE | ID: mdl-37163615

RÉSUMÉ

The rapid emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that evade immunity elicited by vaccination has placed an imperative on the development of countermeasures that provide broad protection against SARS-CoV-2 and related sarbecoviruses. Here, we identified extremely potent monoclonal antibodies (mAbs) that neutralized multiple sarbecoviruses from macaques vaccinated with AS03-adjuvanted monovalent subunit vaccines. Longitudinal analysis revealed progressive accumulation of somatic mutation in the immunoglobulin genes of antigen-specific memory B cells (MBCs) for at least 1 year after primary vaccination. Antibodies generated from these antigen-specific MBCs at 5 to 12 months after vaccination displayed greater potency and breadth relative to those identified at 1.4 months. Fifteen of the 338 (about 4.4%) antibodies isolated at 1.4 to 6 months after the primary vaccination showed potency against SARS-CoV-2 BA.1, despite the absence of serum BA.1 neutralization. 25F9 and 20A7 neutralized authentic clade 1 sarbecoviruses (SARS-CoV, WIV-1, SHC014, SARS-CoV-2 D614G, BA.1, and Pangolin-GD) and vesicular stomatitis virus-pseudotyped clade 3 sarbecoviruses (BtKY72 and PRD-0038). 20A7 and 27A12 showed potent neutralization against all SARS-CoV-2 variants and multiple Omicron sublineages, including BA.1, BA.2, BA.3, BA.4/5, BQ.1, BQ.1.1, and XBB. Crystallography studies revealed the molecular basis of broad and potent neutralization through targeting conserved sites within the RBD. Prophylactic protection of 25F9, 20A7, and 27A12 was confirmed in mice, and administration of 25F9 particularly provided complete protection against SARS-CoV-2, BA.1, SARS-CoV, and SHC014 challenge. These data underscore the extremely potent and broad activity of these mAbs against sarbecoviruses.


Sujet(s)
COVID-19 , Virus du SRAS , Animaux , Humains , Souris , Anticorps neutralisants à large spectre , Vaccins contre la COVID-19 , Macaca , SARS-CoV-2 , COVID-19/prévention et contrôle , Immunisation , Vaccination , Anticorps monoclonaux , Anticorps antiviraux , Anticorps neutralisants
12.
Immunity ; 56(3): 669-686.e7, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36889306

RÉSUMÉ

Pan-betacoronavirus neutralizing antibodies may hold the key to developing broadly protective vaccines against novel pandemic coronaviruses and to more effectively respond to SARS-CoV-2 variants. The emergence of Omicron and subvariants of SARS-CoV-2 illustrates the limitations of solely targeting the receptor-binding domain (RBD) of the spike (S) protein. Here, we isolated a large panel of broadly neutralizing antibodies (bnAbs) from SARS-CoV-2 recovered-vaccinated donors, which targets a conserved S2 region in the betacoronavirus spike fusion machinery. Select bnAbs showed broad in vivo protection against all three deadly betacoronaviruses, SARS-CoV-1, SARS-CoV-2, and MERS-CoV, which have spilled over into humans in the past two decades. Structural studies of these bnAbs delineated the molecular basis for their broad reactivity and revealed common antibody features targetable by broad vaccination strategies. These bnAbs provide new insights and opportunities for antibody-based interventions and for developing pan-betacoronavirus vaccines.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , Anticorps neutralisants à large spectre , Anticorps neutralisants , Anticorps antiviraux
13.
Nat Commun ; 13(1): 7864, 2022 12 21.
Article de Anglais | MEDLINE | ID: mdl-36543789

RÉSUMÉ

Contemporary influenza A H3N2 viruses circulating since 2016 have acquired a glycosylation site in the neuraminidase in close proximity to the enzymatic active site. Here, we investigate if this S245N glycosylation site, as a result of antigenic evolution, can impact binding and function of human monoclonal antibodies that target the conserved active site. While we find that a reduction in the inhibitory ability of neuraminidase active site binders is measurable, this class of broadly reactive monoclonal antibodies maintains protective efficacy in vivo.


Sujet(s)
Anticorps monoclonaux , Sous-type H3N2 du virus de la grippe A , Sialidase , Humains , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/immunologie , Anticorps antiviraux/composition chimique , Anticorps antiviraux/métabolisme , Domaine catalytique/immunologie , Domaine catalytique/physiologie , Glycosylation , Glycoprotéine hémagglutinine du virus influenza , Virus de la grippe A , Sous-type H3N2 du virus de la grippe A/immunologie , Sous-type H3N2 du virus de la grippe A/métabolisme , Grippe humaine/immunologie , Grippe humaine/métabolisme , Sialidase/composition chimique , Sialidase/immunologie
14.
Sci Transl Med ; 14(657): eabl9605, 2022 08 10.
Article de Anglais | MEDLINE | ID: mdl-35947674

RÉSUMÉ

To prepare for future coronavirus (CoV) pandemics, it is desirable to generate vaccines capable of eliciting broadly neutralizing antibody responses to CoVs. Here, we show that immunization of macaques with SARS-CoV-2 spike (S) protein with a two-shot protocol generated potent serum receptor binding domain cross-neutralizing antibody responses to both SARS-CoV-2 and SARS-CoV-1. Furthermore, responses were equally effective against most SARS-CoV-2 variants of concern (VOCs) and some were highly effective against Omicron. This result contrasts with human infection or many two-shot vaccination protocols where responses were typically more SARS-CoV-2 specific and where VOCs were less well neutralized. Structural studies showed that cloned macaque neutralizing antibodies, particularly using a given heavy chain germline gene, recognized a relatively conserved region proximal to the angiotensin converting enzyme 2 receptor binding site (RBS), whereas many frequently elicited human neutralizing antibodies targeted more variable epitopes overlapping the RBS. B cell repertoire differences between humans and macaques appeared to influence the vaccine response. The macaque neutralizing antibodies identified a pan-SARS-related virus epitope region less well targeted by human antibodies that could be exploited in rational vaccine design.


Sujet(s)
COVID-19 , SARS-CoV-2 , Animaux , Anticorps neutralisants , Anticorps antiviraux , Anticorps neutralisants à large spectre , Épitopes , Humains , Macaca mulatta , Glycoprotéine de spicule des coronavirus
15.
Proc Natl Acad Sci U S A ; 119(29): e2205784119, 2022 07 19.
Article de Anglais | MEDLINE | ID: mdl-35767670

RÉSUMÉ

Many neutralizing antibodies (nAbs) elicited to ancestral severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) through natural infection and vaccination have reduced effectiveness to SARS-CoV-2 variants. Here, we show that therapeutic antibody ADG20 is able to neutralize SARS-CoV-2 variants of concern (VOCs) including Omicron (B.1.1.529) as well as other SARS-related coronaviruses. We delineate the structural basis of this relatively escape-resistant epitope that extends from one end of the receptor binding site (RBS) into the highly conserved CR3022 site. ADG20 can then benefit from high potency through direct competition with ACE2 in the more variable RBS and interaction with the more highly conserved CR3022 site. Importantly, antibodies that are able to target this site generally neutralize a broad range of VOCs, albeit with reduced potency against Omicron. Thus, this conserved and vulnerable site can be exploited for the design of universal vaccines and therapeutic antibodies.


Sujet(s)
Anticorps monoclonaux , Anticorps neutralisants , Anticorps antiviraux , COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , COVID-19/virologie , Épitopes/immunologie , Humains , Tests de neutralisation , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie
16.
Proc Natl Acad Sci U S A ; 119(21): e2200821119, 2022 05 24.
Article de Anglais | MEDLINE | ID: mdl-35594401

RÉSUMÉ

Influenza virus hemagglutinin (HA) has been the primary target for influenza vaccine development. Broadly protective antibodies targeting conserved regions of the HA unlock the possibility of generating universal influenza immunity. Two group 2 influenza A chimeric HAs, cH4/3 and cH15/3, were previously designed to elicit antibodies to the conserved HA stem. Here, we show by X-ray crystallography and negative-stain electron microscopy that a broadly protective antistem antibody can stably bind to cH4/3 and cH15/3 HAs, thereby validating their potential as universal vaccine immunogens. Furthermore, flexibility was observed in the head domain of the chimeric HA structures, suggesting that antibodies could also potentially interact with the head interface epitope. Our structural and binding studies demonstrated that a broadly protective antihead trimeric interface antibody could indeed target the more open head domain of the cH15/3 HA trimer. Thus, in addition to inducing broadly protective antibodies against the conserved HA stem, chimeric HAs may also be able to elicit antibodies against the conserved trimer interface in the HA head domain, thereby increasing the vaccine efficacy.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Infections à Orthomyxoviridae , Anticorps neutralisants , Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Hémagglutinines , Humains , Grippe humaine/prévention et contrôle , Infections à Orthomyxoviridae/prévention et contrôle
17.
bioRxiv ; 2022 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-35313576

RÉSUMÉ

Many neutralizing antibodies (nAbs) elicited to ancestral SARS-CoV-2 through natural infection and vaccination generally have reduced effectiveness to SARS-CoV-2 variants. Here we show therapeutic antibody ADG20 is able to neutralize all SARS-CoV-2 variants of concern (VOCs) including Omicron (B.1.1.529) as well as other SARS-related coronaviruses. We delineate the structural basis of this relatively escape-resistant epitope that extends from one end of the receptor binding site (RBS) into the highly conserved CR3022 site. ADG20 can then benefit from high potency through direct competition with ACE2 in the more variable RBS and interaction with the more highly conserved CR3022 site. Importantly, antibodies that are able to target this site generally neutralize all VOCs, albeit with reduced potency against Omicron. Thus, this highly conserved and vulnerable site can be exploited for design of universal vaccines and therapeutic antibodies.

18.
Sci Transl Med ; 14(637): eabi9215, 2022 03 23.
Article de Anglais | MEDLINE | ID: mdl-35133175

RÉSUMÉ

Broadly neutralizing antibodies (bnAbs) to coronaviruses (CoVs) are valuable in their own right as prophylactic and therapeutic reagents to treat diverse CoVs and as templates for rational pan-CoV vaccine design. We recently described a bnAb, CC40.8, from a CoV disease 2019 (COVID-19) convalescent donor that exhibits broad reactivity with human ß-CoVs. Here, we showed that CC40.8 targets the conserved S2 stem helix region of the CoV spike fusion machinery. We determined a crystal structure of CC40.8 Fab with a SARS-CoV-2 S2 stem peptide at 1.6-Å resolution and found that the peptide adopted a mainly helical structure. Conserved residues in ß-CoVs interacted with CC40.8 antibody, thereby providing a molecular basis for its broad reactivity. CC40.8 exhibited in vivo protective efficacy against SARS-CoV-2 challenge in two animal models. In both models, CC40.8-treated animals exhibited less weight loss and reduced lung viral titers compared to controls. Furthermore, we noted that CC40.8-like bnAbs are relatively rare in human COVID-19 infection, and therefore, their elicitation may require rational structure-based vaccine design strategies. Overall, our study describes a target on ß-CoV spike proteins for protective antibodies that may facilitate the development of pan-ß-CoV vaccines.


Sujet(s)
COVID-19 , Glycoprotéine de spicule des coronavirus , Animaux , Anticorps neutralisants/métabolisme , Anticorps antiviraux , COVID-19/immunologie , Humains , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie
19.
J Virol ; 96(5): e0212021, 2022 03 09.
Article de Anglais | MEDLINE | ID: mdl-35044215

RÉSUMÉ

Influenza A viruses (IAV) initiate infection by binding to glycans with terminal sialic acids on the cell surface. Hosts of IAV variably express two major forms of sialic acid, N-acetylneuraminic acid (NeuAc) and N-glycolylneuraminic acid (NeuGc). NeuGc is produced in most mammals, including horses and pigs, but is absent in humans, ferrets, and birds. The only known naturally occurring IAV that exclusively bind NeuGc are extinct highly pathogenic equine H7N7 viruses. We determined the crystal structure of a representative equine H7 hemagglutinin (HA) in complex with NeuGc and observed high similarity in the receptor-binding domain with an avian H7 HA. To determine the molecular basis for NeuAc and NeuGc specificity, we performed systematic mutational analyses, based on the structural insights, on two distant avian H7 HAs and an H15 HA. We found that the A135E mutation is key for binding α2,3-linked NeuGc but does not abolish NeuAc binding. The additional mutations S128T, I130V, T189A, and K193R converted the specificity from NeuAc to NeuGc. We investigated the residues at positions 128, 130, 135, 189, and 193 in a phylogenetic analysis of avian and equine H7 HAs. This analysis revealed a clear distinction between equine and avian residues. The highest variability was observed at key position 135, of which only the equine glutamic acid led to NeuGc binding. These results demonstrate that genetically distinct H7 and H15 HAs can be switched from NeuAc to NeuGc binding and vice versa after the introduction of several mutations, providing insights into the adaptation of H7 viruses to NeuGc receptors. IMPORTANCE Influenza A viruses cause millions of cases of severe illness and deaths annually. To initiate infection and replicate, the virus first needs to bind to a structure on the cell surface, like a key fitting in a lock. For influenza A viruses, these "keys" (receptors) on the cell surface are chains of sugar molecules (glycans). The terminal sugar on these glycans is often either N-acetylneuraminic acid (NeuAc) or N-glycolylneuraminic acid (NeuGc). Most influenza A viruses bind NeuAc, but a small minority bind NeuGc. NeuGc is present in species like horses, pigs, and mice but not in humans, ferrets, and birds. Here, we investigated the molecular determinants of NeuGc specificity and the origin of viruses that bind NeuGc.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza , Sous-type H7N7 du virus de la grippe A , Acides neuraminiques , Animaux , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Equus caballus , Humains , Sous-type H7N7 du virus de la grippe A/composition chimique , Sous-type H7N7 du virus de la grippe A/métabolisme , Acide N-acétyl-neuraminique , Acides neuraminiques/composition chimique , Acides neuraminiques/métabolisme , Phylogenèse , Polyosides/métabolisme , Liaison aux protéines
20.
bioRxiv ; 2022 Jan 21.
Article de Anglais | MEDLINE | ID: mdl-33821273

RÉSUMÉ

Broadly neutralizing antibodies (bnAbs) to coronaviruses (CoVs) are valuable in their own right as prophylactic and therapeutic reagents to treat diverse CoVs and, importantly, as templates for rational pan-CoV vaccine design. We recently described a bnAb, CC40.8, from a coronavirus disease 2019 (COVID-19)-convalescent donor that exhibits broad reactivity with human beta-coronaviruses (ß-CoVs). Here, we showed that CC40.8 targets the conserved S2 stem-helix region of the coronavirus spike fusion machinery. We determined a crystal structure of CC40.8 Fab with a SARS-CoV-2 S2 stem-peptide at 1.6 Å resolution and found that the peptide adopted a mainly helical structure. Conserved residues in ß-CoVs interacted with CC40.8 antibody, thereby providing a molecular basis for its broad reactivity. CC40.8 exhibited in vivo protective efficacy against SARS-CoV-2 challenge in two animal models. In both models, CC40.8-treated animals exhibited less weight loss and reduced lung viral titers compared to controls. Furthermore, we noted CC40.8-like bnAbs are relatively rare in human COVID-19 infection and therefore their elicitation may require rational structure-based vaccine design strategies. Overall, our study describes a target on ß-CoV spike proteins for protective antibodies that may facilitate the development of pan-ß-CoV vaccines. SUMMARY: A human mAb isolated from a COVID-19 donor defines a protective cross-neutralizing epitope for pan-ß-CoV vaccine design strategies.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...