Research paper
High efficiency human memory B cell assay and its application to studying Plasmodium falciparum-specific memory B cells in natural infections

https://doi.org/10.1016/j.jim.2011.09.006Get rights and content

Abstract

Memory B cells (MBCs) are a key component of long term humoral immunity to many human infectious diseases. Despite their importance, we know little about the generation or maintenance of antigen-(Ag)-specific MBCs in humans in response to infection. A frequently employed method for quantifying Ag-specific MBCs in human peripheral blood (Crotty et al., 2004) relies on the ability of MBCs but not naïve B cells to differentiate into antibody secreting cells (ASCs) in response to polyclonal activators and Toll-like receptor agonists in vitro and the measurement of Ag-specific ASCs by ELISPOT assays. Here we report on studies to optimize the efficiency of this ELISPOT-based assay and to apply this assay to the detection of Plasmodium falciparum (Pf)-specific MBCs in adults living in a malaria endemic area where immunity to Pf is acquired through natural infection. We show that the addition of IL-10 to in vitro cultures of human peripheral blood mononuclear cells increased the efficiency of the assay from 10% to over 90% without increasing the ASC burst size and without any substantial increase in background from naïve B cells or plasma cells (PCs). Using this assay we were able to quantify the frequency of Pf-specific MBCs in peripheral blood of adults living in a malaria endemic area. Thus, this highly efficient assay appears to be well suited to field studies of the generation and maintenance of MBCs where the volumes of blood obtainable are often limiting.

Introduction

A hallmark of adaptive immunity is Ag-specific immunological memory, the ability to respond more rapidly and robustly to re-exposure to an Ag. Indeed, all current vaccines are predicated on the phenomenon of immunological memory (Sallusto et al., 2010). However, despite its importance we still have an incomplete understanding of the cellular and molecular mechanisms that underlie the generation, maintenance and activation of immunological memory. For many infectious diseases neutralizing antibodies (Abs) play a critical role in protective immune responses (Sallusto et al., 2010), and thus the mechanisms that underlie the generation and maintenance of B cell memory are of considerable interest. Of particular interest is an understanding of the acquisition of B cell memory in infectious diseases for which we currently have no vaccines, including malaria.

Long-term humoral memory is encoded in both memory B cells (MBCs) and long-lived plasma cells (LLPCs) that are generated during a primary immune response to vaccination or infection. LLPCs reside in the bone marrow and are responsible for maintaining serum antibody (Ab) levels in the absence of Ag re-exposure (Radbruch et al., 2006, Wrammert et al., 2009). Because of their inaccessibility to routine sampling in humans, LLPCs are rarely directly assayed. However, the presence of Ag-specific Ab in serum in the absence of re-exposure to an Ag is likely an accurate reflection of the presence of Ag-specific LLPCs. MBCs are responsible for the rapid, high titer, high affinity secondary Ab responses elicited upon re-exposure to a pathogen or booster vaccination (Radbruch et al., 2006, Wrammert et al., 2009). MBCs reside both in the lymphoid tissues and in the peripheral circulation. The results of a recent study in humans receiving a smallpox vaccine provided evidence that although the majority of vaccine-specific MBCs were in the spleen, the frequency of Ag-specific MBCs in the spleen paralleled the frequency observed in peripheral blood throughout the course of the response (Blink et al., 2005). Furthermore, a recent study using a mouse model of malaria indicated MBCs and PCs in peripheral blood mononuclear cells (PBMCs) reflect Plasmodium-specific B cell responses in the spleen and bone marrow (Nduati et al., 2010). Based on these observations, the frequencies of MBCs in peripheral blood appear to be a good indication of the total number of MBCs in an individual.

Here we focus on the development of B cell memory to Pf malaria because it kills nearly one million people annually and there is no vaccine. Immunity to clinical malaria is slow to develop requiring years of repeated infections (Langhorne et al., 2008, Crompton et al., 2010, Weiss et al., 2010). Abs are known to play a central role in protection from malaria (Cohen et al., 1961) but the B cell biology that underlies the slow acquisition of immunity is just beginning to be investigated. Therefore it is important to develop highly efficient assays that provide the tools for assessing MBCs in the PBMCs of individuals acquiring malaria immunity through natural infection. Evidence is accumulating that the immune systems of individuals living in malaria endemic areas may differ from those in non-malaria endemic areas. For example, in one study we determined that individuals in Mali, in an area of high malaria transmission, have greatly expanded populations of atypical or ‘exhausted’ MBCs (Weiss et al., 2009). In another, we found that Malian adults were relatively refractory to the potentiating effects of CpG-containing vaccines as compared to individuals in the U.S. in that while CpG-containing vaccines resulted in a significant increase in the number of MBCs and the Ab titer in the U.S., there was no difference in MBC number or antibody titer when CpG-containing or non-CpG-containing vaccines were administered to Malian adults (Crompton et al., 2009). Thus, it is essential that assays to detect Pf-specific MBCs are validated in field samples from individuals living in malaria endemic areas.

An assay frequently used to detect Ag-specific human MBCs, described by Crotty et al., 2004 (Crotty et al., 2004), relies on the selective ability of MBCs to proliferate and differentiate into Ab secreting cells (ASCs) in vitro in response to a combination of pokeweed mitogen (PWM), fixed S. aureus, Cowan strain (SAC) and the TLR9 agonist CpG oligonucleotide (ODN-2006) over a five to six day culture period. Ag-specific and total ASCs were quantified in ELISPOT assays using plates coated with either Ag or human Ig-specific antibodies to capture Ag-specific and total IgG, respectively. Crotty et al. showed that peripheral blood MBCs (defined as CD19+CD20+CD27+) from individuals immunized with anthrax vaccine differentiated into anthrax protective antigen (PA)-specific ASCs in this assay, but naïve B cells (defined as CD19+CD20+CD27) did not. PA-specific MBCs represented up to 2% of circulating IgG+ B cells in immune individuals and were essentially undetectable in non-immune individuals. Thus, this assay provided a means of identifying Ag-specific MBCs in human peripheral blood.

Using peripheral blood samples from vaccinated U.S. individuals we determined the efficiency of this assay by enumerating the actual number of tetanus toxoid (TT)-specific MBCs in a given sample using a sensitive and specific flow cytometry technique (Amanna and Slifka, 2006) and comparing this number to the number of MBCs which respond to the stimulation cocktail used by Crotty et al. We report that only ~ 10% of TT-specific MBCs enumerated by flow cytometry respond in a limiting dilution version of the in vitro assay described by Crotty et al. and that the efficiency of this assay could be significantly improved, to nearly 100%, by the addition of IL-10 to the five day culture. This increase in efficiency in a method that can be done with whole PBMCs and does not require isolation of B cells as do other highly efficient assays (Amanna and Slifka, 2006) makes this protocol valuable in field studies in malaria endemic areas, where isolation of B cells is impractical due to both sample number and sample size. We provide evidence that this modified assay can be successfully used to quantify Pf-specific MBCs in semi-immune adults in Kenya. The increased efficiency of this modified assay allows for the measurement of MBCs from smaller blood volumes than was possible with the original assay that should facilitate field studies, particularly those involving children.

Section snippets

Samples and sample preparation

Peripheral blood was obtained from healthy, anonymous, adults at the National Institutes of Health blood bank and peripheral blood mononuclear cells (PBMCs) were isolated from whole blood or elutriated mononuclear cell buffy coats obtained by lymphapheresis within 6 h of blood collection, using by density gradient centrifugation. Samples were diluted in an equal volume of PBS, layered over a 20% volume of Ficoll–Hypaque (Amersham Biosciences) and centrifuged for 20 min at room temperature at 1800 ×

Quantifying antigen-specific MBCs by flow cytometry

To quantify Ag-specific MBCs in vaccinated U.S. individuals we used a sensitive flow cytometry-based assay described for detecting TT-specific MBCs by Amanna and Slifka (Amanna and Slifka, 2006). We chose to measure TT for the assessment of the MBC ELISPOT assay because our attempts to detect Pf-specific MBCs using the Ags apical membrane antigen 1 (AMA1) and merozoite surface protein 1 (MSP1), were unsuccessful, because of the instability and non-specific binding of these proteins. PBMCs were

References (20)

  • I.J. Amanna et al.

    Quantitation of rare memory B cell populations by two independent and complementary approaches

    J. Immunol. Methods

    (2006)
  • S. Crotty et al.

    Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system

    J. Immunol. Methods

    (2004)
  • F. Sallusto et al.

    From vaccines to memory and back

    Immunity

    (2010)
  • I.J. Amanna et al.

    Duration of humoral immunity to common viral and vaccine antigens

    N. Engl. J. Med.

    (2007)
  • C. Arpin et al.

    Generation of memory B cells and plasma cells in vitro

    Science

    (1995)
  • C. Arpin et al.

    Memory B cells are biased towards terminal differentiation: a strategy that may prevent repertoire freezing

    J. Exp. Med.

    (1997)
  • E.J. Blink et al.

    Early appearance of germinal center-derived memory B cells and plasma cells in blood after primary immunization

    J. Exp. Med.

    (2005)
  • J. Choe et al.

    IL-10 interrupts memory B cell expansion in the germinal center by inducing differentiation into plasma cells

    Eur. J. Immunol.

    (1998)
  • S. Cohen et al.

    Gamma-globulin and acquired immunity to human malaria

    Nature

    (1961)
  • P.D. Crompton et al.

    The TLR9 ligand CpG promotes the acquisition of Plasmodium falciparum-specific memory B cells in malaria-naive individuals

    J. Immunol.

    (2009)
There are more references available in the full text version of this article.

Cited by (26)

  • Somatically Hypermutated Plasmodium-Specific IgM<sup>+</sup> Memory B Cells Are Rapid, Plastic, Early Responders upon Malaria Rechallenge

    2016, Immunity
    Citation Excerpt :

    The persistence of heterogeneous MBCs after malaria infection in mice led us to ask whether switched and unswitched P. falciparum-specific MBCs also occur in exposed individuals residing in an endemic area. Although IgG+ P. falciparum-specific MBCs have been detected by ELISPOT (Weiss et al., 2012) in individuals exposed to both high (Ndungu et al., 2012; Weiss et al., 2010) and low (Clark et al., 2012; Ndungu et al., 2013; Wipasa et al., 2010) malaria transmission, it is unknown whether P. falciparum infection induces antigen-specific unswitched MBCs. Antigen-specific enrichment experiments were therefore performed on peripheral blood mononuclear cells (PBMCs) collected from P. falciparum-infected Malian subjects during the malaria season (Crompton et al., 2008) or malaria-naive U.S. subjects.

  • Classification of common variable immunodeficiencies using flow cytometry and a memory B-cell functionality assay

    2015, Journal of Allergy and Clinical Immunology
    Citation Excerpt :

    PBMCs were flow cytometrically analyzed before and after culture under standard incubation settings for 6 days in 6-well plates at 3e6 cells/3 mL complete RPMI with the stimulation cocktail.35 In brief, we used 6 μg/mL of the Toll-like receptor 9 ligand CpG,36 1:10,000 Staphylococcus aureus Cowan activating Toll-like receptor 2, 1 ng/mL pokeweed mitogen with a great effect on B-cell proliferation and T-cell activation,37 and 50 μmol/L β-mercaptoethanol stimulating B- and T-cell help, according to the method of Crotty et al,35 to achieve optimal antigen-unspecific stimulation.38-40 ELISpot assay was performed with standardized 96-well plates commercially coated by our partner, GenID (Straßberg, Germany), to detect total IgG, IgM, and IgA secretion at several concentrations per well (2.5 × 105 ex vivo and 1.25 × 104, 6.5 × 103, 3.1 × 103, and 1.5 × 103 in vitro).

  • Optimization of a human IgG B-cell ELISpot assay for the analysis of vaccine-induced B-cell responses

    2013, Journal of Immunological Methods
    Citation Excerpt :

    Another aspect of the B-cell ELISpot protocol is the kinetics of the memory B-cell activation. Generally, cells are pre-stimulated for 5–6 days prior to being added to the ELISpot plate for the quantification of ASC (Crotty et al., 2004; Buisman et al., 2009; Cao et al., 2010; Weiss et al., 2012). The duration of the stimulation is likely to depend on the potency of the activation.

View all citing articles on Scopus
View full text