Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Nature ; 584(7821): 450-456, 2020 08.
Article in English | MEDLINE | ID: mdl-32698192

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) pandemic continues, with devasting consequences for human lives and the global economy1,2. The discovery and development of virus-neutralizing monoclonal antibodies could be one approach to treat or prevent infection by this coronavirus. Here we report the isolation of sixty-one SARS-CoV-2-neutralizing monoclonal antibodies from five patients infected with SARS-CoV-2 and admitted to hospital with severe coronavirus disease 2019 (COVID-19). Among these are nineteen antibodies that potently neutralized authentic SARS-CoV-2 in vitro, nine of which exhibited very high potency, with 50% virus-inhibitory concentrations of 0.7 to 9 ng ml-1. Epitope mapping showed that this collection of nineteen antibodies was about equally divided between those directed against the receptor-binding domain (RBD) and those directed against the N-terminal domain (NTD), indicating that both of these regions at the top of the viral spike are immunogenic. In addition, two other powerful neutralizing antibodies recognized quaternary epitopes that overlap with the domains at the top of the spike. Cryo-electron microscopy reconstructions of one antibody that targets the RBD, a second that targets the NTD, and a third that bridges two separate RBDs showed that the antibodies recognize the closed, 'all RBD-down' conformation of the spike. Several of these monoclonal antibodies are promising candidates for clinical development as potential therapeutic and/or prophylactic agents against SARS-CoV-2.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Epitopes, B-Lymphocyte/immunology , Pneumonia, Viral/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/ultrastructure , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/analysis , Antibodies, Viral/chemistry , Antibodies, Viral/ultrastructure , Betacoronavirus/chemistry , Betacoronavirus/ultrastructure , COVID-19 , Coronavirus Infections/prevention & control , Cryoelectron Microscopy , Disease Models, Animal , Epitope Mapping , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/ultrastructure , Female , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/ultrastructure , Lung/pathology , Lung/virology , Male , Mesocricetus , Models, Molecular , Neutralization Tests , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/ultrastructure
2.
Sci Rep ; 10(1): 5350, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210344

ABSTRACT

The major barrier to a HIV-1 cure is the persistence of latent genomes despite treatment with antiretrovirals. To investigate host factors which promote HIV-1 latency, we conducted a genome-wide functional knockout screen using CRISPR-Cas9 in a HIV-1 latency cell line model. This screen identified IWS1, POLE3, POLR1B, PSMD1, and TGM2 as potential regulators of HIV-1 latency, of which PSMD1 and TMG2 could be confirmed pharmacologically. Further investigation of PSMD1 revealed that an interacting enzyme, the deubiquitinase UCH37, was also involved in HIV-1 latency. We therefore conducted a comprehensive evaluation of the deubiquitinase family by gene knockout, identifying several deubiquitinases, UCH37, USP14, OTULIN, and USP5 as possible HIV-1 latency regulators. A specific inhibitor of USP14, IU1, reversed HIV-1 latency and displayed synergistic effects with other latency reversal agents. IU1 caused degradation of TDP-43, a negative regulator of HIV-1 transcription. Collectively, this study is the first comprehensive evaluation of deubiquitinases in HIV-1 latency and establishes that they may hold a critical role.


Subject(s)
CRISPR-Cas Systems , Deubiquitinating Enzymes/genetics , Gene Knockout Techniques/methods , HIV-1/physiology , Virus Latency , DNA Polymerase III/genetics , DNA-Binding Proteins/genetics , DNA-Directed RNA Polymerases/genetics , Deubiquitinating Enzyme CYLD/genetics , Deubiquitinating Enzymes/antagonists & inhibitors , Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , HIV-1/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Jurkat Cells , Nucleoproteins/genetics , Proteasome Endopeptidase Complex/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Ubiquitin Thiolesterase/genetics , Virus Latency/drug effects
3.
bioRxiv ; 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32587975

ABSTRACT

The SARS-CoV-2 pandemic rages on with devasting consequences on human lives and the global economy 1,2 . The discovery and development of virus-neutralizing monoclonal antibodies could be one approach to treat or prevent infection by this novel coronavirus. Here we report the isolation of 61 SARS-CoV-2-neutralizing monoclonal antibodies from 5 infected patients hospitalized with severe disease. Among these are 19 antibodies that potently neutralized the authentic SARS-CoV-2 in vitro , 9 of which exhibited exquisite potency, with 50% virus-inhibitory concentrations of 0.7 to 9 ng/mL. Epitope mapping showed this collection of 19 antibodies to be about equally divided between those directed to the receptor-binding domain (RBD) and those to the N-terminal domain (NTD), indicating that both of these regions at the top of the viral spike are immunogenic. In addition, two other powerful neutralizing antibodies recognized quaternary epitopes that are overlapping with the domains at the top of the spike. Cryo-electron microscopy reconstructions of one antibody targeting RBD, a second targeting NTD, and a third bridging two separate RBDs revealed recognition of the closed, "all RBD-down" conformation of the spike. Several of these monoclonal antibodies are promising candidates for clinical development as potential therapeutic and/or prophylactic agents against SARS-CoV-2.

4.
Immunol Lett ; 188: 46-52, 2017 08.
Article in English | MEDLINE | ID: mdl-28610800

ABSTRACT

Humanized mouse models present an important tool for preclinical evaluation of new vaccines and therapeutics. Here we show the human variable repertoire of antibody sequences cloned from a previously described human immune system (HIS) mouse model that possesses functional human CD4+ T cells and B cells, namely HIS-CD4/B mice. We sequenced variable IgG genes from single memory B-cell and plasma-cell sorted from splenocytes or whole blood lymphocytes of HIS-CD4/B mice that were vaccinated with a human plasmodial antigen, a recombinant Plasmodium falciparum circumsporozoite protein (rPfCSP). We demonstrate that rPfCSP immunization triggers a diverse B-cell IgG repertoire composed of various human VH family genes and distinct V(D)J recombinations that constitute diverse CDR3 sequences similar to humans, although low hypermutated sequences were generated. These results demonstrate the substantial genetic diversity of responding human B cells of HIS-CD4/B mice and their capacity to mount human IgG class-switched antibody response upon vaccination.


Subject(s)
Antigens, Protozoan/immunology , Immune System/immunology , Immunoglobulin G/immunology , Malaria/immunology , Plasmodium/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Gene Amplification , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Heterografts , Humans , Immunoglobulin G/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Malaria/parasitology , Mice , Mice, Transgenic , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410104

ABSTRACT

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Subject(s)
Disease Models, Animal , Immunity, Humoral/immunology , Malaria, Falciparum/immunology , Mice, Transgenic/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Heterografts , Histocompatibility Antigens Class II , Humans , Malaria Vaccines , Mice , Protozoan Proteins/immunology
6.
J Acquir Immune Defic Syndr ; 66(2): 140-7, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24457632

ABSTRACT

BACKGROUND: To understand whether combination antiretroviral therapy (cART) has been optimized, we asked whether 3-drug protease inhibitor (PI)-based cART intensified with raltegravir and maraviroc and initiated during early infection would improve outcomes when compared with similarly applied 3-drug PI-based cART. METHODS: Forty newly HIV-1-infected patients were randomized 1:2 to receive 3-drug (N = 14) or 5-drug (N = 26) therapy. The primary end point was the percent of subjects with undetectable plasma viremia using standard reverse transcriptase-polymerase chain reaction and the single copy assay after 48 weeks. Secondary end points included levels of cell-associated HIV-1 DNA and RNA and levels of infectious virus in resting CD4 T cells at week 96 and quantitative and qualitative immunologic responses. RESULTS: At 48 weeks, 34 subjects remained on study and are included in the as-treated analysis. Three of 11 (27.3%) in the 3-drug arm and 9 of 21 (42.9%) in the 5-drug arm had plasma HIV-1 RNA levels below detection by both standard reverse transcriptase-polymerase chain reaction and single copy assay (P = 0.46, Fisher exact test). No significant differences in absolute levels of proviral DNA or changes in cell-associated RNA were seen during 96 weeks of therapy. Mean levels of infectious HIV-1 in resting CD4 T cells at week 96 in 7 subjects treated with 3-drugs and 13 with 5-drugs were 0.67 and 0.71 infectious units per million, respectively (P = 0.81). No differences were seen in quantitative or qualitative immunologic determinations including markers of immune activation. CONCLUSIONS: Intensified 5-drug cART initiated during early infection fails to significantly further impact virologic or immunologic responses beyond those achieved with standard 3-drug PI-based cART.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/methods , HIV Infections/drug therapy , Adult , Aged , CD4 Lymphocyte Count , Cyclohexanes/therapeutic use , DNA, Viral/blood , Drug Combinations , Endpoint Determination , HIV Infections/blood , Humans , Longitudinal Studies , Male , Maraviroc , Middle Aged , Pyrrolidinones/therapeutic use , RNA, Viral/blood , Raltegravir Potassium , Triazoles/therapeutic use , Viral Load , Viremia/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL