Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pathogens ; 12(5)2023 May 02.
Article in English | MEDLINE | ID: mdl-37242341

ABSTRACT

A multiplexed enzyme-linked immunosorbent assay (ELISA) that simultaneously measures antibody binding to multiple antigens can extend the impact of serosurveillance studies, particularly if the assay approaches the simplicity, robustness, and accuracy of a conventional single-antigen ELISA. Here, we report on the development of multiSero, an open-source multiplex ELISA platform for measuring antibody responses to viral infection. Our assay consists of three parts: (1) an ELISA against an array of proteins in a 96-well format; (2) automated imaging of each well of the ELISA array using an open-source plate reader; and (3) automated measurement of optical densities for each protein within the array using an open-source analysis pipeline. We validated the platform by comparing antibody binding to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) antigens in 217 human sera samples, showing high sensitivity (0.978), specificity (0.977), positive predictive value (0.978), and negative predictive value (0.977) for classifying seropositivity, a high correlation of multiSero determined antibody titers with commercially available SARS-CoV-2 antibody tests, and antigen-specific changes in antibody titer dynamics upon vaccination. The open-source format and accessibility of our multiSero platform can contribute to the adoption of multiplexed ELISA arrays for serosurveillance studies, for SARS-CoV-2 and other pathogens of significance.

2.
PLoS Comput Biol ; 18(9): e1010052, 2022 09.
Article in English | MEDLINE | ID: mdl-36126074

ABSTRACT

The sequencing of antibody repertoires of B-cells at increasing coverage and depth has led to the identification of vast numbers of immunoglobulin heavy and light chains. However, the size and complexity of these Adaptive Immune Receptor Repertoire sequencing (AIRR-seq) datasets makes it difficult to perform exploratory analyses. To aid in data exploration, we have developed AIRRscape, an R Shiny-based interactive web browser application that enables B-cell receptor (BCR) and antibody feature discovery through comparisons among multiple repertoires. Using AIRR-seq data as input, AIRRscape starts by aggregating and sorting repertoires into interactive and explorable bins of germline V-gene, germline J-gene, and CDR3 length, providing a high-level view of the entire repertoire. Interesting subsets of repertoires can be quickly identified and selected, and then network topologies of CDR3 motifs can be generated for further exploration. Here we demonstrate AIRRscape using patient BCR repertoires and sequences of published monoclonal antibodies to investigate patterns of humoral immunity to three viral pathogens: SARS-CoV-2, HIV-1, and DENV (dengue virus). AIRRscape reveals convergent antibody sequences among datasets for all three pathogens, although HIV-1 antibody datasets display limited convergence and idiosyncratic responses. We have made AIRRscape available as a web-based Shiny application, along with code on GitHub to encourage its open development and use by immuno-informaticians, virologists, immunologists, vaccine developers, and other scientists that are interested in exploring and comparing multiple immune receptor repertoires.


Subject(s)
Antibody Formation , COVID-19 , Antibodies, Monoclonal , High-Throughput Nucleotide Sequencing , Humans , Receptors, Antigen, B-Cell/genetics , SARS-CoV-2/genetics
3.
medRxiv ; 2021 May 11.
Article in English | MEDLINE | ID: mdl-34013298

ABSTRACT

Serology has provided valuable diagnostic and epidemiological data on antibody responses to SARS-CoV-2 in diverse patient cohorts. Deployment of high content, multiplex serology platforms across the world, including in low and medium income countries, can accelerate longitudinal epidemiological surveys. Here we report multiSero, an open platform to enable multiplex serology with up to 48 antigens in a 96-well format. The platform consists of three components: ELISA-array of printed proteins, a commercial or home-built plate reader, and modular python software for automated analysis (pysero). We validate the platform by comparing antibody titers against the SARS-CoV-2 Spike, receptor binding domain (RBD), and nucleocapsid (N) in 114 sera from COVID-19 positive individuals and 87 pre-pandemic COVID-19 negative sera. We report data with both a commercial plate reader and an inexpensive, open plate reader (nautilus). Receiver operating characteristic (ROC) analysis of classification with single antigens shows that Spike and RBD classify positive and negative sera with the highest sensitivity at a given specificity. The platform distinguished positive sera from negative sera when the reactivity of the sera was equivalent to the binding of 1 ng mL âˆ'1 RBD-specific monoclonal antibody. We developed normalization and classification methods to pool antibody responses from multiple antigens and multiple experiments. Our results demonstrate a performant and accessible pipeline for multiplexed ELISA ready for multiple applications, including serosurveillance, identification of viral proteins that elicit antibody responses, differential diagnosis of circulating pathogens, and immune responses to vaccines.

4.
J Immunol Methods ; 481-482: 112789, 2020.
Article in English | MEDLINE | ID: mdl-32380014

ABSTRACT

We describe an adaptation of conventional ELISA methods to an ELISA-Array format using non-contact Piezo printing of up to 30 spots of purified recombinant viral fusion proteins and vaccine on 96 well high-protein binding plates. Antigens were printed in 1 nanoliter volumes of protein stabilizing buffer using as little as 0.25 nanograms of protein, 2000-fold less than conventional ELISA. The performance of the ELISA-Array was demonstrated by serially diluting n = 9 human post-flu vaccination plasma samples starting at a 1/1000 dilution and measuring binding to the array of Influenza antigens. Plasma polyclonal antibody levels were detected using a cocktail of biotinylated anti-human kappa and lambda light chain antibodies, followed by a Streptavidin-horseradish peroxidase conjugate and the dose-dependent signal was developed with a precipitable TMB substrate. Intra- and inter-assay precision of absorbance units among the eight donor samples showed mean CVs of 4.8% and 10.8%, respectively. The plasma could be differentiated by donor and antigen with titer sensitivities ranging from 1 × 103 to 4 × 106, IC50 values from 1 × 104 to 9 × 106, and monoclonal antibody sensitivities in the ng/mL range. Equivalent sensitivities of ELISA versus ELISA-Array, compared using plasma and an H1N1 HA trimer, were achieved on the ELISA-Array printed at 0.25 ng per 200um spot and 1000 ng per ELISA 96-well. Vacuum-sealed array plates were shown to be stable when stored for at least 2 days at ambient temperature and up to 1 month at 4-8 °C. By the use of any set of printed antigens and analyte matrices the methods of this multiplexed ELISA-Array format can be broadly applied in translational research.


Subject(s)
Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/blood , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/blood
5.
Elife ; 82019 12 10.
Article in English | MEDLINE | ID: mdl-31820734

ABSTRACT

Eliciting broadly neutralizing antibodies (bNAbs) against the four dengue virus serotypes (DENV1-4) that are spreading into new territories is an important goal of vaccine design. To define bNAb targets, we characterized 28 antibodies belonging to expanded and hypermutated clonal families identified by transcriptomic analysis of single plasmablasts from DENV-infected individuals. Among these, we identified J9 and J8, two somatically related bNAbs that potently neutralized DENV1-4. Mutagenesis studies showed that the major recognition determinants of these bNAbs are in E protein domain I, distinct from the only known class of human bNAbs against DENV with a well-defined epitope. B cell repertoire analysis from acute-phase peripheral blood suggested that J9 and J8 followed divergent somatic hypermutation pathways, and that a limited number of mutations was sufficient for neutralizing activity. Our study suggests multiple B cell evolutionary pathways leading to DENV bNAbs targeting a new epitope that can be exploited for vaccine design.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Dengue Virus/immunology , Dengue/immunology , Gene Expression Profiling , Antibodies, Neutralizing/genetics , Antibodies, Viral/genetics , DNA Mutational Analysis , Humans , Protein Binding , Viral Envelope Proteins/metabolism
6.
Front Immunol ; 10: 1452, 2019.
Article in English | MEDLINE | ID: mdl-31293598

ABSTRACT

Phenotypic screening of antigen-specific antibodies in human blood is a common diagnostic test for infectious agents and a correlate of protection after vaccination. In addition to long-lived antibody secreting plasma cells residing in the bone marrow, memory B cells are a latent source of antigen-experienced, long-term immunity that can be found at low frequencies in circulating peripheral blood mononuclear cells (PBMCs). Assessing the genotype, clonal frequency, quality, and function of antibodies resulting from an individual's persistent memory B cell repertoire can help inform the success or failure of immune protection. Using in vitro polyclonal stimulation, we functionally expand the memory repertoire from PBMCs and clonally map monoclonal antibodies from this population. We show that combining deep sequencing of stimulated memory B cell repertoires with retrieving single antigen-specific cells is a promising approach in evaluating the latent, functional B cell memory in PBMCs.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Plasma Cells/immunology , Antibody Formation/immunology , Cell Line , Cells, Cultured , Clone Cells/immunology , Humans , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Leukocytes, Mononuclear/immunology
7.
MAbs ; 6(2): 460-73, 2014.
Article in English | MEDLINE | ID: mdl-24492306

ABSTRACT

Viral entry targets with therapeutic neutralizing potential are subject to multiple escape mechanisms, including antigenic drift, immune dominance of functionally irrelevant epitopes, and subtle variations in host cell mechanisms. A surprising finding of recent years is that potent neutralizing antibodies to viral epitopes independent of strain exist, but are poorly represented across the diverse human population. Identifying these antibodies and understanding the biology mediating the specific immune response is thus difficult. An effective strategy for meeting this challenge is to incorporate multiplexed antigen screening into a high throughput survey of the memory B cell repertoire from immune individuals. We used this approach to discover suites of cross-clade antibodies directed to conformational epitopes in the stalk region of the influenza A hemagglutinin (HA) protein and to select high-affinity anti-peptide antibodies to the glycoprotein B (gB) of human cytomegalovirus. In each case, our screens revealed a restricted VH and VL germline usage, including published and previously unidentified gene families. The in vivo evolution of paratope specificity with optimal neutralizing activity was understandable after correlating biological activities with kinetic binding and epitope recognition. Iterative feedback between antigen probe design based on structure and function information with high throughput multiplexed screening demonstrated a generally applicable strategy for efficient identification of safe, native, finely tuned antibodies with the potential for high genetic barriers to viral escape.


Subject(s)
Antibodies, Blocking/metabolism , Antigens, Viral/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Epitopes/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/immunology , Influenza, Human/immunology , Viral Envelope Proteins/metabolism , Antibodies, Blocking/immunology , Antibody Affinity , Antigens, Viral/immunology , Cell Line , Cytomegalovirus Infections/therapy , Hemagglutinin Glycoproteins, Influenza Virus/immunology , High-Throughput Screening Assays , Humans , Immune Evasion/drug effects , Immunity, Humoral , Immunity, Innate , Immunologic Memory , Influenza, Human/therapy , Protein Conformation , Viral Envelope Proteins/immunology
8.
PLoS One ; 7(3): e33106, 2012.
Article in English | MEDLINE | ID: mdl-22427961

ABSTRACT

Graft-versus-host disease (GVHD) is a major barrier to successful allogeneic hematopoietic cell transplantation and is largely mediated by activated donor lymphocytes. Lymphotoxin (LT)-α is expressed by subsets of activated T and B cells, and studies in preclinical models demonstrated that targeted depletion of these cells with a mouse anti-LT-α monoclonal antibody (mAb) was efficacious in inhibiting inflammation and autoimmune disease. Here we demonstrate that LT-α is also upregulated on activated human donor lymphocytes in a xenogeneic model of GVHD and targeted depletion of these donor cells ameliorated GVHD. A depleting humanized anti-LT-α mAb, designated MLTA3698A, was generated that specifically binds to LT-α in both the soluble and membrane-bound forms, and elicits antibody-dependent cellular cytotoxicity (ADCC) activity in vitro. Using a human peripheral blood mononuclear cell transplanted SCID (Hu-SCID) mouse model of GVHD, the anti-human LT-α mAb specifically depleted activated LT-expressing human donor T and B cells, resulting in prolonged survival of the mice. A mutation in the Fc region, rendering the mAb incapable of mediating ADCC, abolished all in vitro and in vivo effects. These data support a role for using a depleting anti-LT-α antibody in treating immune diseases such as GVHD and autoimmune diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Lymphocytes/immunology , Lymphotoxin-alpha/deficiency , Transplantation, Homologous/adverse effects , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lymphotoxin-alpha/immunology , Mice , Mice, SCID , Surface Plasmon Resonance , Transplantation, Homologous/immunology
9.
J Immunol Methods ; 358(1-2): 35-45, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20361968

ABSTRACT

We developed a neutralizing antibody assay (NAb assay), based upon the complement-dependent cytotoxicity (CDC) activity of a monoclonal human IgG(1) therapeutic (IgT), to characterize anti-therapeutic antibodies (ATA) in autoimmune patient serum. Neutralizing antibodies (NAb) were measured by a decrease in the extent of CDC mediated by 50 ng/mL IgT, on a lymphoblastoid cell line. A sample pre-treatment procedure, utilizing a Protein A/G resin to purify total immunoglobulins, was optimized for use in the NAb assay to eliminate the non-specific assay interferents observed in individual serum samples from rheumatoid arthritis patients. In some individuals, the addition of naïve serum to the assay completely inhibited CDC activity. After sample pre-treatment, the variability of the CDC response induced by IgT in individual serum samples from a drug-naïve RA population, tested over three days, was only 3%, irrespective of complement immune complexes or rheumatoid factor levels. The pre-treatment procedure was performed on samples and matrix controls as part of each assay. The NAb assay was able to recover and detect polyclonal ATA from human serum at a concentration of 0.25 microg/mL with pre-treatment. Dose-dependent neutralization of IgT was observed, however, a simple positive/negative reporting scheme was adopted. The NAb assay was found to have the desired properties of specificity, robustness, precision and recovery for validation to support the characterization of ATA in clinical samples.


Subject(s)
Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Artifacts , Cytotoxicity Tests, Immunologic/methods , Immunoassay/methods , Serum/chemistry , Serum/immunology , Animals , Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , Cell Line, Tumor , Complement Pathway, Classical/immunology , Cross Reactions/immunology , Goats/immunology , Humans , Immunoglobulin G/immunology , Macaca fascicularis/immunology , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...