skip to main content


Title: Analysis of Antigen‐Specific Human Memory B Cell Populations Based on In Vitro Polyclonal Stimulation
Abstract

Antigen‐specific memory B cell (MBC) populations mediate the rapid, strong, and high‐affinity secondary antibody responses that play a key role in combating infection and generating protective responses to vaccination. Recently, cell staining with fluorochrome‐labeled antigens together with sequencing methods such as Drop‐seq and CITE‐seq have provided information on the specificity, phenotype, and transcriptome of single MBCs. However, characterization of MBCs at the level of antigen‐reactive populations remains an important tool for assessing an individual's B cell immunity and responses to antigen exposure. This is readily performed using a long‐established method based on in vitro polyclonal stimulation of MBCs to induce division and differentiation into antibody‐secreting cells (ASCs). Post‐stimulation antigen‐specific measurement of the MBC‐derived ASCs (or the secreted antibodies) indicates the size of precursor MBC populations. Additional information about the character of antigen‐reactive MBC populations is provided by analysis of MBC‐derived antibodies of particular specificities for binding avidity and functionality. This article outlines a simple and reliable strategy for efficient in vitro MBC stimulation and use of the ELISpot assay as a post‐stimulation readout to determine the size of antigen‐specific MBC populations. Other applications of the in vitro stimulation technique for MBC analysis are discussed. The following protocols are included. © 2020 Wiley Periodicals LLC

Basic Protocol 1: Polyclonal stimulation of memory B cells using unfractionated PBMCs

Alternate Protocol: Stimulation of small PBMC numbers using 96‐well plates with U‐bottom wells

Basic Protocol 2: ELISpot assay for enumeration of memory B cell−derived antibody‐secreting cells

 
more » « less
NSF-PAR ID:
10238193
Author(s) / Creator(s):
 ;  ;  ;  
Publisher / Repository:
Wiley Blackwell (John Wiley & Sons)
Date Published:
Journal Name:
Current Protocols in Immunology
Volume:
131
Issue:
1
ISSN:
1934-3671
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Abstract

    In this invited article, we explain technical aspects of the lymphocytic choriomeningitis virus (LCMV) system, providing an update of a prior contribution by Matthias von Herrath and J. Lindsay Whitton. We provide an explanation of the LCMV infection models, highlighting the importance of selecting an appropriate route and viral strain. We also describe how to quantify virus‐specific immune responses, followed by an explanation of useful transgenic systems. Specifically, our article will focus on the following protocols. © 2020 Wiley Periodicals LLC.

    Basic Protocol 1: LCMV infection routes in mice

    Support Protocol 1: Preparation of LCMV stocks

    ASSAYS TO MEASURE LCMV TITERS

    Support Protocol 2: Plaque assay

    Support Protocol 3: Immunofluorescence focus assay (IFA) to measure LCMV titer

    MEASUREMENT OF T CELL AND B CELL RESPONSES TO LCMV INFECTION

    Basic Protocol 2: Triple tetramer staining for detection of LCMV‐specific CD8 T cells

    Basic Protocol 3: Intracellular cytokine staining (ICS) for detection of LCMV‐specific T cells

    Basic Protocol 4: Enumeration of direct ex vivo LCMV‐specific antibody‐secreting cells (ASC)

    Basic Protocol 5: Limiting dilution assay (LDA) for detection of LCMV‐specific memory B cells

    Basic Protocol 6: ELISA for quantification of LCMV‐specific IgG antibody

    Support Protocol 4: Preparation of splenic lymphocytes

    Support Protocol 5: Making BHK21‐LCMV lysate

    Basic Protocol 7: Challenge models

    TRANSGENIC MODELS

    Basic Protocol 8: Transfer of P14 cells to interrogate the role of IFN‐I on CD8 T cell responses

    Basic Protocol 9: Comparing the expansion of naïve versus memory CD4 T cells following chronic viral challenge

     
    more » « less
  2. Abstract

    Isolating cells based on their secreted proteins remain a challenge. The authors demonstrate a capacity for high throughput single‐cell protein secretion analysis and isolation based on heterofunctional particles combined with fluorescence activated cell sorting (FACS). The workflow shows that antibody secreting cells (ASCs) specific for the H1 protein from influenza virus can be isolated from B cells. The workflow consists of incubating anti‐CD27 particles with the ASCs, capturing locally secreted immunoglobulins with Protein G on the particles, and identifying immunoglobulins specific to H1 via fluorescent labeled antigens followed by FACS to enrich antigen‐specific ASCs. Two particles designs, Janus and mixed, are tested with hybridoma cells. Mixed particles are found to improve antibody collection, while Janus particles are found to bind target cells more effectively. Targeted hybridoma cells in coculture with non‐specific hybridoma cells are identified with a sensitivity of 96% and specificity of 98%. Heterofunctional particles are used to capture ASCs that secrete antibodies specific for influenza virus from B cells from healthy adults isolated from blood after vaccination. Positive H1‐tetramer sorted ASCs are validated using single ASC cultures and identify 23/56 cells specific for H1 demonstrating 164‐fold enrichment from total B cells and 14.6‐fold enrichment from total ASCs.

     
    more » « less
  3. Abstract

    Cross‐presentation was first observed serendipitously in the 1970s. The importance of it was quickly realized and subsequently attracted great attention from immunologists. Since then, our knowledge of the ability of certain antigen presenting cells to internalize, process, and load exogenous antigens onto MHC‐I molecules to cross‐prime CD8+T cells has increased significantly. Dendritic cells (DCs) are exceptional cross‐presenters, thus making them a great tool to study cross‐presentation but the relative rarity of DCs in circulation and in tissues makes it challenging to isolate sufficient numbers of cells to study this process in vitro. In this paper, we describe in detail two methods to culture DCs from bone‐marrow progenitors and a method to expand the numbers of DCs present in vivo as a source of endogenous bona‐fide cross‐presenting DCs. We also describe methods to assess cross‐presentation by DCs using the activation of primary CD8+T cells as a readout. © 2020 Wiley Periodicals LLC.

    Basic Protocol 1: Isolation of bone marrow progenitor cells

    Basic Protocol 2: In vitro differentiation of dendritic cells with GM‐CSF

    Support Protocol 1: Preparation of conditioned medium from GM‐CSF producing J558L cells

    Basic Protocol 3: In vitro differentiation of dendritic cells with Flt3L

    Support Protocol 2: Preparation of Flt3L containing medium from B16‐Flt3L cells

    Basic Protocol 4: Expansion of cDC1s in vivo for use in ex vivo experiments

    Basic Protocol 5: Characterizing resting and activated dendritic cells

    Basic Protocol 6: Dendritic cell stimulation, antigenic cargo, and fixation

    Support Protocol 3: Preparation of model antigen coated microbeads

    Support Protocol 4: Preparation of apoptotic cells

    Support Protocol 5: Preparation of recombinant bacteria

    Basic Protocol 7: Immunocytochemistry immunofluorescence (ICC/IF)

    Support Protocol 6: Preparation of Alcian blue‐coated coverslips

    Basic Protocol 8: CD8+T cell activation to assess cross‐presentation

    Support Protocol 7: Isolation and labeling of CD8+T cells with CFSE

     
    more » « less
  4. Abstract

    Diet‐induced obesity is associated with impaired B‐cell‐driven humoral immunity, which coincides with chronic inflammation and has consequences for responses to infections and vaccinations. Key nutritional, cellular, and molecular mechanisms by which obesity may impair aspects of humoral immunity such as B cell development, class switch recombination, and formation of long‐lived antibody secreting cells are reviewed. A key theme to emerge is the central role of white adipose tissue on the formation and function of pro‐inflammatory B cell subsets that exacerbate insulin resistance. The underlying role of select hormones such as leptin is highlighted, which may be driving the formation of pro‐inflammatory B cells in the absence of antigen stimulation. This review also extensively covers the regulatory role of lipid metabolites such as prostaglandins and specialized pro‐resolving mediators (SPMs) that are synthesized from polyunsaturated fatty acids. Notably, SPM biosynthesis is impaired in obesity and contributes toward impaired antibody production. Future directions for research, including avenues for therapeutic intervention, are included.

     
    more » « less
  5. Abstract

    Extracellular vesicles (EVs) are sub‐micron‐sized membranous spheres secreted by cells. EVs play a functional role as intercellular communicators and are associated with a number of diseases. Research into EVs is an area of growing interest due their many potential uses as therapeutic agents, as diagnostic and theranostic biomarkers, and as regulators of cellular biology. Flow cytometry is a popular method for enumerating and phenotyping EVs, even though the majority of EVs are below the detection sensitivity of most commercially available flow cytometers. Here, we present optimized protocols for EV labeling that increase the signal‐to‐noise ratio of EVs by removing residual antibody. Protocols for alignment of high‐resolution jet‐in‐air flow cytometers are also provided. Published 2020. U.S. Government.

    Basic Protocol 1: Bulk EV staining with CFSE protein binding dye

    Basic Protocol 2: Antigen‐specific staining of EV markers with fluorochrome‐conjugated antibodies

    Basic Protocol 3: Astrios EQ instrument setup and sample acquisition

    Basic Protocol 4: Counting particles and EVs on Astrios EQ with spike‐in reference beads

     
    more » « less