Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
bioRxiv ; 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38903070

RÉSUMÉ

Broadly neutralizing antibodies targeting the V2 apex of the HIV-1 envelope trimer are among the most common specificities elicited in HIV-1-infected humans and simian-human immunodeficiency virus (SHIV)-infected macaques. To gain insight into the prevalent induction of these antibodies, we isolated and characterized 11 V2 apex-directed neutralizing antibody lineages from SHIV-infected rhesus macaques. Remarkably, all SHIV-induced V2 apex lineages were derived from reading frame two of the rhesus DH3-15*01 gene. Cryo-EM structures of envelope trimers in complex with antibodies from nine rhesus lineages revealed modes of recognition that mimicked three canonical human V2 apex-recognition modes. Notably, amino acids encoded by DH3-15*01 played divergent structural roles, inserting into a hole at the trimer apex, H-bonding to an exposed strand, or forming part of a loop scaffold. Overall, we identify a DH3-15*01-signature for rhesus V2 apex broadly neutralizing antibodies and show that highly selected genetic elements can play multiple roles in antigen recognition. Highlights: Isolated 11 V2 apex-targeted HIV-neutralizing lineages from 10 SHIV-infected Indian-origin rhesus macaquesCryo-EM structures of Fab-Env complexes for nine rhesus lineages reveal modes of recognition that mimic three modes of human V2 apex antibody recognitionAll SHIV-elicited V2 apex lineages, including two others previously published, derive from the same DH3-15*01 gene utilizing reading frame twoThe DH3-15*01 gene in reading frame two provides a necessary, but not sufficient, signature for V2 apex-directed broadly neutralizing antibodiesStructural roles played by DH3-15*01-encoded amino acids differed substantially in different lineages, even for those with the same recognition modePropose that the anionic, aromatic, and extended character of DH3-15*01 in reading frame two provides a selective advantage for V2 apex recognition compared to B cells derived from other D genes in the naïve rhesus repertoireDemonstrate that highly selected genetic elements can play multiple roles in antigen recognition, providing a structural means to enhance recognition diversity.

2.
Cell Rep ; 43(3): 113948, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38483908

RÉSUMÉ

Identifying individual functional B cell receptors (BCRs) is common, but two-dimensional analysis of B cell frequency versus BCR potency would delineate both quantity and quality of antigen-specific memory B cells. We efficiently determine quantitative BCR neutralizing activities using a single-cell-derived antibody supernatant analysis (SCAN) workflow and develop a frequency-potency algorithm to estimate B cell frequencies at various neutralizing activity or binding affinity cutoffs. In an HIV-1 fusion peptide (FP) immunization study, frequency-potency curves elucidate the quantity and quality of FP-specific immunoglobulin G (IgG)+ memory B cells for different animals, time points, and antibody lineages at single-cell resolution. The BCR neutralizing activities are mainly determined by their affinities to soluble envelope trimer. Frequency analysis definitively demonstrates dominant neutralizing antibody lineages. These findings establish SCAN and frequency-potency analyses as promising approaches for general B cell analysis and monoclonal antibody (mAb) discovery. They also provide specific rationales for HIV-1 FP-directed vaccine optimization.


Sujet(s)
Infections à VIH , Séropositivité VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Animaux , Anticorps neutralisants , Anticorps anti-VIH , Immunoglobuline G , Cellules B mémoire
3.
Nat Med ; 29(10): 2535-2546, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37783968

RÉSUMÉ

The main barrier to HIV cure is a persistent reservoir of latently infected CD4+ T cells harboring replication-competent provirus that fuels rebound viremia upon antiretroviral therapy (ART) interruption. A leading approach to target this reservoir involves agents that reactivate latent HIV proviruses followed by direct clearance of cells expressing induced viral antigens by immune effector cells and immunotherapeutics. We previously showed that AZD5582, an antagonist of inhibitor of apoptosis proteins and mimetic of the second mitochondrial-derived activator of caspases (IAPi/SMACm), induces systemic reversal of HIV/SIV latency but with no reduction in size of the viral reservoir. In this study, we investigated the effects of AZD5582 in combination with four SIV Env-specific Rhesus monoclonal antibodies (RhmAbs) ± N-803 (an IL-15 superagonist) in SIV-infected, ART-suppressed rhesus macaques. Here we confirm the efficacy of AZD5582 in inducing SIV reactivation, demonstrate enhancement of latency reversal when AZD5582 is used in combination with N-803 and show a reduction in total and replication-competent SIV-DNA in lymph-node-derived CD4+ T cells in macaques treated with AZD5582 + RhmAbs. Further exploration of this therapeutic approach may contribute to the goal of achieving an HIV cure.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Virus de l'immunodéficience simienne/physiologie , Macaca mulatta , Antirétroviraux/pharmacologie , Antirétroviraux/usage thérapeutique , Latence virale , Réplication virale , Anticorps/usage thérapeutique , Noeuds lymphatiques , Lymphocytes T CD4+ , Charge virale
4.
Cell Rep ; 42(8): 112942, 2023 08 29.
Article de Anglais | MEDLINE | ID: mdl-37561630

RÉSUMÉ

Zika virus (ZIKV) is an emerging pathogen that causes devastating congenital defects. The overlapping epidemiology and immunologic cross-reactivity between ZIKV and dengue virus (DENV) pose complex challenges to vaccine design, given the potential for antibody-dependent enhancement of disease. Therefore, classification of ZIKV-specific antibody targets is of notable value. From a ZIKV-infected rhesus macaque, we identify ZIKV-reactive B cells and isolate potent neutralizing monoclonal antibodies (mAbs) with no cross-reactivity to DENV. We group these mAbs into four distinct antigenic groups targeting ZIKV-specific cross-protomer epitopes on the envelope glycoprotein. Co-crystal structures of representative mAbs in complex with ZIKV envelope glycoprotein reveal envelope-dimer epitope and unique dimer-dimer epitope targeting. All four specificities are serologically identified in convalescent humans following ZIKV infection, and representative mAbs from all four groups protect against ZIKV replication in mice. These results provide key insights into ZIKV-specific antigenicity and have implications for ZIKV vaccine, diagnostic, and therapeutic development.


Sujet(s)
Virus de la dengue , Dengue , Vaccins antiviraux , Infection par le virus Zika , Virus Zika , Humains , Animaux , Souris , Anticorps neutralisants , Épitopes , Macaca mulatta , Anticorps antiviraux , Anticorps monoclonaux , Vaccins antiviraux/usage thérapeutique , Protéines de l'enveloppe virale/composition chimique
5.
J Virol ; 97(4): e0186422, 2023 04 27.
Article de Anglais | MEDLINE | ID: mdl-36976017

RÉSUMÉ

The monoclonal antibodies (MAbs) NCI05 and NCI09, isolated from a vaccinated macaque that was protected from multiple simian immunodeficiency virus (SIV) challenges, both target an overlapping, conformationally dynamic epitope in SIV envelope variable region 2 (V2). Here, we show that NCI05 recognizes a CH59-like coil/helical epitope, whereas NCI09 recognizes a ß-hairpin linear epitope. In vitro, NCI05 and, to a lesser extent, NCI09 mediate the killing of SIV-infected cells in a CD4-dependent manner. Compared to NCI05, NCI09 mediates higher titers of antibody-dependent cellular cytotoxicity (ADCC) to gp120-coated cells, as well as higher levels of trogocytosis, a monocyte function that contributes to immune evasion. We also found that passive administration of NCI05 or NCI09 to macaques did not affect the risk of SIVmac251 acquisition compared to controls, demonstrating that these anti-V2 antibodies alone are not protective. However, NCI05 but not NCI09 mucosal levels strongly correlated with delayed SIVmac251 acquisition, and functional and structural data suggest that NCI05 targets a transient state of the viral spike apex that is partially opened, compared to its prefusion-closed conformation. IMPORTANCE Studies suggest that the protection against SIV/simian-human immunodeficiency virus (SHIV) acquisition afforded by the SIV/HIV V1 deletion-containing envelope immunogens, delivered by the DNA/ALVAC vaccine platform, requires multiple innate and adaptive host responses. Anti-inflammatory macrophages and tolerogenic dendritic cells (DC-10), together with CD14+ efferocytes, are consistently found to correlate with a vaccine-induced decrease in the risk of SIV/SHIV acquisition. Similarly, V2-specific antibody responses mediating ADCC, Th1 and Th2 cells expressing no or low levels of CCR5, and envelope-specific NKp44+ cells producing interleukin 17 (IL-17) also are reproducible correlates of decreased risk of virus acquisition. We focused on the function and the antiviral potential of two monoclonal antibodies (NCI05 and NCI09) isolated from vaccinated animals that differ in antiviral function in vitro and recognize V2 in a linear (NCI09) or coil/helical (NCI05) conformation. We demonstrate that NCI05, but not NCI09, delays SIVmac251 acquisition, highlighting the complexity of antibody responses to V2.


Sujet(s)
Anticorps monoclonaux , Virus de l'immunodéficience simienne , Protéines virales , Virus de l'immunodéficience simienne/immunologie , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/isolement et purification , Anticorps monoclonaux/métabolisme , Protéines virales/composition chimique , Protéines virales/immunologie , Épitopes/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Structure tertiaire des protéines , Modèles moléculaires , Cellules CHO , Cricetulus , Animaux , Macaca/immunologie , Macaca/virologie , Anticorps antiviraux/sang
6.
Nat Struct Mol Biol ; 29(11): 1080-1091, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36344847

RÉSUMÉ

Simian immunodeficiency viruses (SIVs) are lentiviruses that naturally infect non-human primates of African origin and seeded cross-species transmissions of HIV-1 and HIV-2. Here we report prefusion stabilization and cryo-EM structures of soluble envelope (Env) trimers from rhesus macaque SIV (SIVmac) in complex with neutralizing antibodies. These structures provide residue-level definition for SIV-specific disulfide-bonded variable loops (V1 and V2), which we used to delineate variable-loop coverage of the Env trimer. The defined variable loops enabled us to investigate assembled Env-glycan shields throughout SIV, which we found to comprise both N- and O-linked glycans, the latter emanating from V1 inserts, which bound the O-link-specific lectin jacalin. We also investigated in situ SIVmac-Env trimers on virions, determining cryo-electron tomography structures at subnanometer resolutions for an antibody-bound complex and a ligand-free state. Collectively, these structures define the prefusion-closed structure of the SIV-Env trimer and delineate variable-loop and glycan-shielding mechanisms of immune evasion conserved throughout SIV evolution.


Sujet(s)
Anticorps neutralisants , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Animaux , Cryomicroscopie électronique , Macaca mulatta/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Tomographie en microscopie électronique , 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
7.
PLoS Pathog ; 18(6): e1010574, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35709309

RÉSUMÉ

Both SIV and SHIV are powerful tools for evaluating antibody-mediated prevention and treatment of HIV-1. However, owing to a lack of rhesus-derived SIV broadly neutralizing antibodies (bnAbs), testing of bnAbs for HIV-1 prevention or treatment has thus far been performed exclusively in the SHIV NHP model using bnAbs from HIV-1-infected individuals. Here we describe the isolation and characterization of multiple rhesus-derived SIV bnAbs capable of neutralizing most isolates of SIV. Eight antibodies belonging to two clonal families, ITS102 and ITS103, which target unique epitopes in the CD4 binding site (CD4bs) region, were found to be broadly neutralizing and together neutralized all SIV strains tested. A rare feature of these bnAbs and two additional antibody families, ITS92 and ITS101, which mediate strain-specific neutralizing activity against SIV from sooty mangabeys (SIVsm), was their ability to achieve near complete (i.e. 100%) neutralization of moderately and highly neutralization-resistant SIV. Overall, these newly identified SIV bnAbs highlight the potential for evaluating HIV-1 prophylactic and therapeutic interventions using fully simian, rhesus-derived bnAbs in the SIV NHP model, thereby circumventing issues related to rapid antibody clearance of human-derived antibodies, Fc mismatch and limited genetic diversity of SHIV compared to SIV.


Sujet(s)
Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Virus de l'immunodéficience simienne , Animaux , Anticorps neutralisants , Anticorps neutralisants à large spectre , Anticorps anti-VIH , Macaca mulatta
8.
Science ; 373(6556)2021 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-34210892

RÉSUMÉ

The emergence of highly transmissible SARS-CoV-2 variants of concern (VOCs) that are resistant to therapeutic antibodies highlights the need for continuing discovery of broadly reactive antibodies. We identified four receptor binding domain-targeting antibodies from three early-outbreak convalescent donors with potent neutralizing activity against 23 variants, including the B.1.1.7, B.1.351, P.1, B.1.429, B.1.526, and B.1.617 VOCs. Two antibodies are ultrapotent, with subnanomolar neutralization titers [half-maximal inhibitory concentration (IC50) 0.3 to 11.1 nanograms per milliliter; IC80 1.5 to 34.5 nanograms per milliliter). We define the structural and functional determinants of binding for all four VOC-targeting antibodies and show that combinations of two antibodies decrease the in vitro generation of escape mutants, suggesting their potential in mitigating resistance development.


Sujet(s)
Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/immunologie , Angiotensin-converting enzyme 2/antagonistes et inhibiteurs , Angiotensin-converting enzyme 2/métabolisme , Anticorps neutralisants/composition chimique , Anticorps neutralisants/métabolisme , Anticorps antiviraux/composition chimique , Anticorps antiviraux/métabolisme , Affinité des anticorps , Réaction antigène-anticorps , COVID-19/virologie , Humains , Échappement immunitaire , Fragments Fab d'immunoglobuline/immunologie , Fragments Fab d'immunoglobuline/métabolisme , Mutation , Tests de neutralisation , Domaines protéiques , Récepteurs du coronavirus/antagonistes et inhibiteurs , Récepteurs du coronavirus/métabolisme , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme
10.
bioRxiv ; 2021 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-33655252

RÉSUMÉ

The emergence of highly transmissible SARS-CoV-2 variants of concern (VOC) that are resistant to therapeutic antibodies highlights the need for continuing discovery of broadly reactive antibodies. We identify four receptor-binding domain targeting antibodies from three early-outbreak convalescent donors with potent neutralizing activity against 12 variants including the B.1.1.7 and B.1.351 VOCs. Two of them are ultrapotent, with sub-nanomolar neutralization titers (IC50 <0.0006 to 0.0102 µ g/mL; IC80 < 0.0006 to 0.0251 µ g/mL). We define the structural and functional determinants of binding for all four VOC-targeting antibodies, and show that combinations of two antibodies decrease the in vitro generation of escape mutants, suggesting potential means to mitigate resistance development. These results define the basis of therapeutic cocktails against VOCs and suggest that targeted boosting of existing immunity may increase vaccine breadth against VOCs.

11.
Science ; 371(6525)2021 01 08.
Article de Anglais | MEDLINE | ID: mdl-33214287

RÉSUMÉ

Neutralizing antibodies elicited by HIV-1 coevolve with viral envelope proteins (Env) in distinctive patterns, in some cases acquiring substantial breadth. We report that primary HIV-1 envelope proteins-when expressed by simian-human immunodeficiency viruses in rhesus macaques-elicited patterns of Env-antibody coevolution very similar to those in humans, including conserved immunogenetic, structural, and chemical solutions to epitope recognition and precise Env-amino acid substitutions, insertions, and deletions leading to virus persistence. The structure of one rhesus antibody, capable of neutralizing 49% of a 208-strain panel, revealed a V2 apex mode of recognition like that of human broadly neutralizing antibodies (bNAbs) PGT145 and PCT64-35S. Another rhesus antibody bound the CD4 binding site by CD4 mimicry, mirroring human bNAbs 8ANC131, CH235, and VRC01. Virus-antibody coevolution in macaques can thus recapitulate developmental features of human bNAbs, thereby guiding HIV-1 immunogen design.


Sujet(s)
Coévolution biologique/immunologie , Anticorps neutralisants à large spectre , Anticorps anti-VIH , Protéine d'enveloppe gp120 du VIH/immunologie , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Sites de fixation , Anticorps neutralisants à large spectre/composition chimique , Anticorps neutralisants à large spectre/génétique , Anticorps neutralisants à large spectre/immunologie , Antigènes CD4/immunologie , Cryomicroscopie électronique , Épitopes/immunologie , Anticorps anti-VIH/composition chimique , Anticorps anti-VIH/génétique , Anticorps anti-VIH/immunologie , Protéine d'enveloppe gp120 du VIH/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Humains , Macaca mulatta , Mimétisme moléculaire/immunologie , Virus de l'immunodéficience simienne/génétique , Réplication virale
12.
Proc Natl Acad Sci U S A ; 117(51): 32566-32573, 2020 12 22.
Article de Anglais | MEDLINE | ID: mdl-33288704

RÉSUMÉ

Acute HIV infection is characterized by rapid viral seeding of immunologic inductive sites in the gut followed by the severe depletion of gut CD4+ T cells. Trafficking of α4ß7-expressing lymphocytes to the gut is mediated by MAdCAM, the natural ligand of α4ß7 that is expressed on gut endothelial cells. MAdCAM signaling through α4ß7 costimulates CD4+ T cells and promotes HIV replication. Similar to MAdCAM, the V2 domain of the gp120 HIV envelope protein binds to α4ß7 In this study, we report that gp120 V2 shares with MAdCAM the capacity to signal through α4ß7 resulting in CD4+ T cell activation and proliferation. As with MAdCAM-mediated costimulation, cellular activation induced by gp120 V2 is inhibited by anti-α4ß7 monoclonal antibodies (mAbs). It is also inhibited by anti-V2 domain antibodies including nonneutralizing mAbs that recognize an epitope in V2 that has been linked to reduced risk of acquisition in the RV144 vaccine trial. The capacity of the V2 domain of gp120 to mediate signaling through α4ß7 likely impacts early events in HIV infection. The capacity of nonneutralizing V2 antibodies to block this activity reveals a previously unrecognized mechanism whereby such antibodies might impact HIV transmission and pathogenesis.


Sujet(s)
Lymphocytes T CD4+/virologie , Protéine d'enveloppe gp120 du VIH/métabolisme , Infections à VIH/métabolisme , Intégrines/métabolisme , Agents antiVIH/immunologie , Agents antiVIH/pharmacologie , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie , Anticorps neutralisants/immunologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/métabolisme , Prolifération cellulaire , Épitopes/immunologie , Épitopes/métabolisme , Protéine d'enveloppe gp120 du VIH/composition chimique , Protéine d'enveloppe gp120 du VIH/immunologie , Infections à VIH/anatomopathologie , Infections à VIH/virologie , Interactions hôte-pathogène/physiologie , Humains , Activation des lymphocytes , Domaines protéiques , Transduction du signal , Virus de l'immunodéficience simienne/immunologie , Trétinoïne/pharmacologie
13.
Immunity ; 53(4): 733-744.e8, 2020 10 13.
Article de Anglais | MEDLINE | ID: mdl-32946741

RÉSUMÉ

Discovering potent human monoclonal antibodies (mAbs) targeting the Plasmodium falciparum circumsporozoite protein (PfCSP) on sporozoites (SPZ) and elucidating their mechanisms of neutralization will facilitate translation for passive prophylaxis and aid next-generation vaccine development. Here, we isolated a neutralizing human mAb, L9 that preferentially bound NVDP minor repeats of PfCSP with high affinity while cross-reacting with NANP major repeats. L9 was more potent than six published neutralizing human PfCSP mAbs at mediating protection against mosquito bite challenge in mice. Isothermal titration calorimetry and multiphoton microscopy showed that L9 and the other most protective mAbs bound PfCSP with two binding events and mediated protection by killing SPZ in the liver and by preventing their egress from sinusoids and traversal of hepatocytes. This study defines the subdominant PfCSP minor repeats as neutralizing epitopes, identifies an in vitro biophysical correlate of SPZ neutralization, and demonstrates that the liver is an important site for antibodies to prevent malaria.


Sujet(s)
Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Anticorps antiprotozoaires/immunologie , Antipaludiques/immunologie , Plasmodium falciparum/immunologie , Protéines de protozoaire/immunologie , Sporozoïtes/immunologie , Adolescent , Adulte , Animaux , Lignée cellulaire , Lignée cellulaire tumorale , Épitopes/immunologie , Femelle , Cellules HEK293 , Hépatocytes/immunologie , Hépatocytes/parasitologie , Humains , Foie/immunologie , Foie/parasitologie , Paludisme/immunologie , Paludisme/parasitologie , Vaccins contre le paludisme/immunologie , Mâle , Souris , Souris de lignée C57BL , Adulte d'âge moyen , Jeune adulte
14.
Immunity ; 51(4): 724-734.e4, 2019 10 15.
Article de Anglais | MEDLINE | ID: mdl-31586542

RÉSUMÉ

HIV- and SIV-envelope (Env) trimers are both extensively glycosylated, and antibodies identified to date have been unable to fully neutralize SIVmac239. Here, we report the isolation, structure, and glycan interactions of antibody ITS90.03, a monoclonal antibody that completely neutralized the highly neutralization-resistant isolate, SIVmac239. The co-crystal structure of a fully glycosylated SIVmac239-gp120 core in complex with rhesus CD4 and the antigen-binding fragment of ITS90.03 at 2.5-Å resolution revealed that ITS90 recognized an epitope comprised of 45% glycan. SIV-gp120 core, rhesus CD4, and their complex could each be aligned structurally to their human counterparts. The structure revealed that glycans masked most of the SIV Env protein surface, with ITS90 targeting a glycan hole, which is occupied in ∼83% of SIV strains by glycan N238. Overall, the SIV glycan shield appears to functionally resemble its HIV counterpart in coverage of spike, shielding from antibody, and modulation of receptor accessibility.


Sujet(s)
Anticorps monoclonaux/composition chimique , Anticorps neutralisants/composition chimique , Infections à VIH/immunologie , VIH (Virus de l'Immunodéficience Humaine)/physiologie , Polyosides/composition chimique , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/physiologie , Animaux , Anticorps monoclonaux/isolement et purification , Anticorps monoclonaux/métabolisme , Anticorps neutralisants/isolement et purification , Anticorps neutralisants/métabolisme , Antigènes CD4/métabolisme , Cellules cultivées , Cristallisation , Cristallographie aux rayons X , Modèles animaux de maladie humaine , Glycosylation , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/métabolisme , Protéine d'enveloppe gp120 du VIH/métabolisme , Humains , Macaca mulatta , Glycoprotéines membranaires/métabolisme , Polyosides/métabolisme , Liaison aux protéines , Relation structure-activité , Protéines de l'enveloppe virale/métabolisme
15.
Science ; 365(6457): 1033-1036, 2019 09 06.
Article de Anglais | MEDLINE | ID: mdl-31488690

RÉSUMÉ

A study in nonhuman primates reported that infusions of an antibody against α4ß7 integrin, in combination with antiretroviral therapy, showed consistent, durable control of simian immunodeficiency virus (SIV) in rhesus macaques. The antibody used has pleiotropic effects, so we set out to gain insight into the underlying mechanism by comparing this treatment to treatment with non-neutralizing monoclonal antibodies against the SIV envelope glycoprotein that only block α4ß7 binding to SIV Env but have no other host-directed effects. Similar to the initial study, we used an attenuated strain of SIV containing a stop codon in nef. The study used 30 macaques that all began antiretroviral therapy and then were divided into five groups to receive different antibody treatments. Unlike the published report, we found no sustained virologic control by these treatments in vivo.


Sujet(s)
Anticorps monoclonaux/usage thérapeutique , Anticorps antiviraux/usage thérapeutique , Intégrine alpha4/immunologie , Chaines bêta des intégrines/immunologie , Syndrome d'immunodéficience acquise du singe/thérapie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/génétique , Virus de l'immunodéficience simienne/immunologie , Animaux , Anticorps monoclonaux/administration et posologie , Anticorps antiviraux/administration et posologie , ADN viral/sang , Produits du gène env/immunologie , Infections à VIH/thérapie , Macaca mulatta , ARN viral/sang , Syndrome d'immunodéficience acquise du singe/sang , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Virus de l'immunodéficience simienne/physiologie , Lymphocytes T/immunologie , Charge virale , Protéines virales régulatrices ou accessoires/génétique , Protéines virales régulatrices ou accessoires/immunologie , Réplication virale
16.
Cell ; 178(3): 567-584.e19, 2019 07 25.
Article de Anglais | MEDLINE | ID: mdl-31348886

RÉSUMÉ

The vaccine-mediated elicitation of antibodies (Abs) capable of neutralizing diverse HIV-1 strains has been a long-standing goal. To understand how broadly neutralizing antibodies (bNAbs) can be elicited, we identified, characterized, and tracked five neutralizing Ab lineages targeting the HIV-1-fusion peptide (FP) in vaccinated macaques over time. Genetic and structural analyses revealed two of these lineages to belong to a reproducible class capable of neutralizing up to 59% of 208 diverse viral strains. B cell analysis indicated each of the five lineages to have been initiated and expanded by FP-carrier priming, with envelope (Env)-trimer boosts inducing cross-reactive neutralization. These Abs had binding-energy hotspots focused on FP, whereas several FP-directed Abs induced by immunization with Env trimer-only were less FP-focused and less broadly neutralizing. Priming with a conserved subregion, such as FP, can thus induce Abs with binding-energy hotspots coincident with the target subregion and capable of broad neutralization.


Sujet(s)
Vaccins contre le SIDA/immunologie , Anticorps neutralisants/immunologie , Anticorps anti-VIH/immunologie , Peptides/immunologie , Séquence d'acides aminés , Animaux , Anticorps neutralisants/composition chimique , Anticorps neutralisants/classification , Lymphocytes B/cytologie , Lymphocytes B/métabolisme , Cristallographie aux rayons X , Femelle , Cellules HEK293 , Anticorps anti-VIH/composition chimique , Anticorps anti-VIH/classification , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Humains , Macaca mulatta , Mâle , Peptides/composition chimique , Structure tertiaire des protéines , Produits du gène env du virus de l'immunodéficience humaine/composition chimique , Produits du gène env du virus de l'immunodéficience humaine/immunologie , Produits du gène env du virus de l'immunodéficience humaine/métabolisme
17.
J Virol ; 93(15)2019 08 01.
Article de Anglais | MEDLINE | ID: mdl-31092584

RÉSUMÉ

We evaluated the contribution of CD8αß+ T cells to control of live-attenuated simian immunodeficiency virus (LASIV) replication during chronic infection and subsequent protection from pathogenic SIV challenge. Unlike previous reports with a CD8α-specific depleting monoclonal antibody (mAb), the CD8ß-specific mAb CD8ß255R1 selectively depleted CD8αß+ T cells without also depleting non-CD8+ T cell populations that express CD8α, such as natural killer (NK) cells and γδ T cells. Following infusion with CD8ß255R1, plasma viremia transiently increased coincident with declining peripheral CD8αß+ T cells. Interestingly, plasma viremia returned to predepletion levels even when peripheral CD8αß+ T cells did not. Although depletion of CD8αß+ T cells in the lymph node (LN) was incomplete, frequencies of these cells were 3-fold lower (P = 0.006) in animals that received CD8ß255R1 than in those that received control IgG. It is possible that these residual SIV-specific CD8αß+ T cells may have contributed to suppression of viremia during chronic infection. We also determined whether infusion of CD8ß255R1 in the LASIV-vaccinated animals increased their susceptibility to infection following intravenous challenge with pathogenic SIVmac239. We found that 7/8 animals infused with CD8ß255R1, and 3/4 animals infused with the control IgG, were resistant to SIVmac239 infection. These results suggest that infusion with CD8ß255R1 did not eliminate the protection afforded to LASIV vaccination. This provides a comprehensive description of the impact of CD8ß255R1 infusion on the immunological composition in cynomolgus macaques, compared to an isotype-matched control IgG, while showing that the control of LASIV viremia and protection from challenge can occur even after CD8ß255R1 administration.IMPORTANCE Studies of SIV-infected macaques that deplete CD8+ T cells in vivo with monoclonal antibodies have provided compelling evidence for their direct antiviral role. These studies utilized CD8α-specific mAbs that target both the major (CD8αß+) and minor (CD8αα+) populations of CD8+ T cells but additionally deplete non-CD8+ T cell populations that express CD8α, such as NK cells and γδ T cells. In the current study, we administered the CD8ß-specific depleting mAb CD8ß255R1 to cynomolgus macaques chronically infected with a LASIV to selectively deplete CD8αß+ T cells without removing CD8αα+ lymphocytes. We evaluated the impact on control of virus replication and protection from pathogenic SIVmac239 challenge. These results underscore the utility of CD8ß255R1 for studying the direct contribution of CD8αß+ T cells in various disease states.


Sujet(s)
Antigènes CD8/analyse , Lymphocytes T CD8+/immunologie , Déplétion lymphocytaire , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Sous-populations de lymphocytes T/immunologie , Réplication virale , Animaux , Macaca , Plasma sanguin/virologie , Virus de l'immunodéficience simienne/croissance et développement , Charge virale
18.
PLoS Pathog ; 14(12): e1007395, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30517201

RÉSUMÉ

Gene based delivery of immunoglobulins promises to safely and durably provide protective immunity to individuals at risk of acquiring infectious diseases such as HIV. We used a rhesus macaque animal model to optimize delivery of naturally-arising, autologous anti-SIV neutralizing antibodies expressed by Adeno-Associated Virus 8 (AAV8) vectors. Vectored transgene expression was confirmed by quantitation of target antibody abundance in serum and mucosal surfaces. We tested the expression achieved at varying doses and numbers of injections. Expression of the transgene reached a saturation at about 2 x 10(12) AAV8 genome copies (gc) per needle-injection, a physical limitation that may not scale clinically into human trials. In contrast, expression increased proportionately with the number of injections. In terms of anti-drug immunity, anti-vector antibody responses were universally strong, while those directed against the natural transgene mAb were detected in only 20% of animals. An anti-transgene antibody response was invariably associated with loss of detectable plasma expression of the antibody. Despite having atypical glycosylation profiles, transgenes derived from AAV-directed muscle cell expression retained full functional activity, including mucosal accumulation, in vitro neutralization, and protection against repeated limiting dose SIVsmE660 swarm challenge. Our findings demonstrate feasibility of a gene therapy-based passive immunization strategy against infectious disease, and illustrate the potential for the nonhuman primate model to inform clinical AAV-based approaches to passive immunization.


Sujet(s)
Anticorps antiviraux/administration et posologie , Thérapie génétique/méthodes , Immunisation passive/méthodes , Vaccins contre le SIDA simien , Protéines de l'enveloppe virale/immunologie , Animaux , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/immunologie , Anticorps neutralisants/administration et posologie , Anticorps neutralisants/immunologie , Dependovirus , Vecteurs génétiques , Macaca mulatta , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne , Transgènes
19.
PLoS Pathog ; 14(8): e1007278, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30153309

RÉSUMÉ

The GI tract is preferentially targeted during acute/early HIV-1 infection. Consequent damage to the gut plays a central role in HIV pathogenesis. The basis for preferential targeting of gut tissues is not well defined. Recombinant proteins and synthetic peptides derived from HIV and SIV gp120 bind directly to integrin α4ß7, a gut-homing receptor. Using both cell-surface expressed α4ß7 and a soluble α4ß7 heterodimer we demonstrate that its specific affinity for gp120 is similar to its affinity for MAdCAM (its natural ligand). The gp120 V2 domain preferentially engages extended forms of α4ß7 in a cation -sensitive manner and is inhibited by soluble MAdCAM. Thus, V2 mimics MAdCAM in the way that it binds to α4ß7, providing HIV a potential mechanism to discriminate between functionally distinct subsets of lymphocytes, including those with gut-homing potential. Furthermore, α4ß7 antagonists developed for the treatment of inflammatory bowel diseases, block V2 binding to α4ß7. A 15-amino acid V2 -derived peptide is sufficient to mediate binding to α4ß7. It includes the canonical LDV/I α4ß7 binding site, a cryptic epitope that lies 7-9 amino acids amino terminal to the LDV/I, and residues K169 and I181. These two residues were identified in a sieve analysis of the RV144 vaccine trial as sites of vaccine -mediated immune pressure. HIV and SIV V2 mAbs elicited by both vaccination and infection that recognize this peptide block V2-α4ß7 interactions. These mAbs recognize conformations absent from the ß- barrel presented in a stabilized HIV SOSIP gp120/41 trimer. The mimicry of MAdCAM-α4ß7 interactions by V2 may influence early events in HIV infection, particularly the rapid seeding of gut tissues, and supports the view that HIV replication in gut tissue is a central feature of HIV pathogenesis.


Sujet(s)
Protéine d'enveloppe gp120 du VIH/composition chimique , Protéine d'enveloppe gp120 du VIH/immunologie , Protéine d'enveloppe gp120 du VIH/métabolisme , Infections à VIH/prévention et contrôle , Intégrines/métabolisme , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Vaccins contre le SIDA/composition chimique , Vaccins contre le SIDA/immunologie , Vaccins contre le SIDA/métabolisme , Animaux , Anticorps monoclonaux , Sites de fixation/immunologie , Lignée cellulaire tumorale , Épitopes/composition chimique , Épitopes/immunologie , Anticorps anti-VIH/composition chimique , Anticorps anti-VIH/immunologie , Anticorps anti-VIH/métabolisme , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Macaca , Liaison aux protéines , Motifs et domaines d'intéraction protéique/immunologie , Vaccins contre le SIDA simien/composition chimique , Vaccins contre le SIDA simien/immunologie , Vaccins contre le SIDA simien/métabolisme , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Vaccination/méthodes
20.
J Infect Dis ; 218(suppl_5): S528-S536, 2018 11 22.
Article de Anglais | MEDLINE | ID: mdl-30010811

RÉSUMÉ

We recently identified a single potently neutralizing monoclonal antibody (mAb), mAb114, isolated from a human survivor of natural Zaire ebolavirus (EBOV) infection, which fully protects nonhuman primates (NHPs) against lethal EBOV challenge. To evaluate the ability of vaccination to generate mAbs such as mAb114, we cloned antibodies from NHPs vaccinated with vectors encoding the EBOV glycoprotein (GP). We identified 14 unique mAbs with potent binding to GP, 4 of which were neutralized and had the functional characteristics of mAb114. These vaccine-induced macaque mAbs share many sequence similarities with mAb114 and use the same mAb114 VH gene (ie, IGHV3-13) when classified using the macaque IMGT database. The antigen-specific VH-gene repertoire present after each immunization indicated that IGHV3-13 mAbs populate an EBOV-specific B-cell repertoire that appears to become more prominent with subsequent boosting. These findings will support structure-based vaccine design aimed at enhanced induction of antibodies such as mAb114.


Sujet(s)
Anticorps monoclonaux/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Lymphocytes B/immunologie , Vaccins contre la maladie à virus Ebola/immunologie , Ebolavirus/immunologie , Animaux , Humains , Concentration en ions d'hydrogène , Macaca fascicularis , Vaccination , Protéines de l'enveloppe virale/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...