Research paper
A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples

https://doi.org/10.1016/j.jim.2010.12.016Get rights and content

Abstract

Phagocytosis can be induced via the engagement of Fcγ receptors by antibody-opsonized material. Furthermore, the efficiency of antibody-induced effector functions has been shown to be dramatically modulated by changes in antibody glycosylation. Because infection can modulate antibody glycans, which in turn modulate antibody functions, assays capable of determining the induction of effector functions rather than neutralization or titer provide a valuable opportunity to more fully characterize the quality of the adaptive immune response. Here we describe a robust and high-throughput flow cytometric assay to define the phagocytic activity of antigen-specific antibodies from clinical samples. This assay employs a monocytic cell line that expresses numerous Fc receptors: including inhibitory and activating, and high and low affinity receptors—allowing complex phenotypes to be studied. We demonstrate the adaptability of this high-throughput, flow-based assay to measure antigen-specific antibody-mediated phagocytosis against an array of viruses, including influenza, HIV, and dengue. The phagocytosis assay format further allows for simultaneous analysis of cytokine release, as well as determination of the role of specific Fcγ-receptor subtypes, making it a highly useful system for parsing differences in the ability of clinical and vaccine induced antibody samples to recruit this critical effector function.

Introduction

Antibodies are potent determinants of the humoral immune response. Though generated as a result of the interaction of B and T cells, antibodies trigger their cytotoxic effects by interacting with complement and innate effector cells. Thus they provide a functional link between the adaptive and innate immune system. They consist of two identical variable domains (Fv) capable of recognizing a target antigen, and a single constant domain (Fc) capable of interacting with the effector cells of the immune system. Traditionally, the epitope recognized by the Fv domains has been thought to be of paramount importance, in that binding to some epitopes can block, or neutralize the native function of the cognate antigen. However, the neutralizing activity mediated by the Fv domains of these antibodies has been found to be insufficient for their protective effects in numerous settings(Clynes et al., 2000, Johnson and Glennie, 2003, Schmidt and Gessner, 2005, Hessell et al., 2007), and evidence of the importance of the constant domain's effector function in clinical outcomes has been accumulating across fields ranging from cancer immunotherapy (Dall'Ozzo et al., 2004) to autoimmunity(Laszlo et al., 1986) and chronic viral infection (Shore et al., 1974). Analogously to the Fv escape mechanisms such as mutating surface epitopes, several pathogens evade the Fc-mediated antibody response by expressing proteases that restrict the Fc domain (Shakirova et al., 1985, Berasain et al., 2000, Collin et al., 2002, Vidarsson et al., 2005, Aslam et al., 2008), or glycosidases that remove the sugar residues required for interaction with Fc receptors (Allhorn et al., 2008). Combined, these evasion mechanisms and clinical correlates provide strong evidence as to the importance of Fc-based effector functions in the therapeutic activity of antibodies.

Significantly, while the primary sequence of the constant domain is conserved across antibodies of a given isotype, the effector functions of distinct antibody isotypes are profoundly modulated by alterations in the glycosylation profile at asparagine 297 (Asn297) in the CH2 domain of the antibody, modulating the range of effector responses a given antibody may elicit (Boyd et al., 1995). The presence or absence of particular sugar groups on the Fc domain tunes the affinity between IgG and Fc receptors (FcγRs) on effector cells, and Fc glycoform represents a potent means of modulating antibody activity (Lund et al., 1996, Raju, 2008). This modulation is bidirectional, as some sugar structures dramatically affect affinity to stimulatory FcRs, while others are known to inhibit immune activation (Shields et al., 2002, Kaneko et al., 2006, Nimmerjahn et al., 2007, Scallon et al., 2007, Raju, 2008, Anthony and Ravetch, 2010).

Similarly, the expression levels of FcγR are also able to modulate antibody activity. Among IgG binding FcRs, multiple isoforms with distinct functions have been identified: FcγgR1 (high affinity, activating), FcγR2a (low affinity, activating), FcγR2b (low affinity, inhibitory), and FcγR3 (low affinity, activating). Thus, FcRs for IgG antibodies include both high and low affinity, as well as activating and inhibitory receptors, each of which may have differential affinities for various IgG glycoforms, and may be expressed at different levels on different cell types. Thus, FcγR expression levels combined with Fc glycosylation patterns represent a highly tunable system for modulating the activity of antibodies.

While numerous classes of innate immune cells express the FcγR involved in antibody-mediated cytotoxicity, a subset of these are capable of acting as professional phagocytes, including monocytes, macrophages, neutrophils, dendritic cells, and mast cells. Phagocytic mechanisms have a demonstrated importance in clearance, antigen presentation, and innate immune activation. Additionally, antibody-driven phagocytosis enhances infection in several infectious diseases (Halstead and O'Rourke, 1977, Marchette et al., 1979, Bernard et al., 1990, Tamura et al., 1991, Fust et al., 1994, Kozlowski et al., 1995) indicating a relevance of phagocytic processes not only on protection from, but in susceptibility to disease, and highlights the significance of this effector function in particular.

Overall, as a potent mechanism of antibody-mediated effector function, antibody-dependent phagocytosis of immune complexes, opsonized pathogens, and host cells represents an important connection between the adaptive and innate immune systems. Because antibody glycosylation patterns and therefore FcR affinity, and immune activity differ, a robust means to measure this critical effector function may help define qualitative differences in this effector function during infection, and post-vaccination that may play an important role in protection. Here we present a high throughput assay capable of measuring the phagocytic activity of antibodies in clinical samples.

Section snippets

Cells, control antibodies, viral envelope antigens

THP-1 cells were purchased from ATCC and cultured as recommended. Care was taken to keep cultures at cell densities below 0.5 × 106/ml in order to maintain consistent levels of FcγR expression and assay performance.

A panel of antibodies to FcγR 1, 2, 2A, 2B, and 3 was used (BD Pharmingen, 558592, 555406; Biolegend, 303212; Novus Biologicals, NB100-79947; RD Systems, AF1875) to determine receptor expression on THP-1 cells. Quantum Simply Cellular bead standards (Bangs Laboratories, 815) were used

The phagocytosis assay

In addition to their role in neutralization, antibodies are able to mediate a number of additional functions including the recruitment of innate immune responses to eliminate antibody-opsonized material. Among these additional antibody mediated functions, antibodies are able to promote phagocytosis, which may play a profound role in the rapid containment and clearance of a pathogen following infection. However, robust assays that are able to capture differences in the quality of

Conclusion

Overall, because innate immune recruiting effector functions have been underappreciated in the protective capacity of even neutralizing antibodies, and phagocytosis might be important in the initial control and clearance of pathogens, an assay capable of measuring this effector function represents an important means of characterizing the immune response to infection or vaccination. Challenges related to the study of effector functions include the large number of FcγRs with variable affinities,

Acknowledgments

The authors wish to acknowledge funding from the International AIDS Vaccine Initiative through the Neutralizing Antibody Consortium; NIAID, NIH AI055332, NIH AI080289, and The Ragon Institute of MGH, MIT, and Harvard. BM acknowledges fellowship support from the Alfred Benzon Foundation. We thank the Phillip T and Susan M Ragon Foundation for their support.

References (49)

  • R.L. Shields et al.

    Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity

    J. Biol. Chem.

    (2002)
  • R.L. Shields et al.

    High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R

    J. Biol. Chem.

    (2001)
  • M. Tamura et al.

    Antibodies to HA and NA augment uptake of influenza A viruses into cells via Fc receptor entry

    Virology

    (1991)
  • Y. Yamaguchi et al.

    Glycoform-dependent conformational alteration of the Fc region of human immunoglobulin G1 as revealed by NMR spectroscopy

    Biochim. Biophys. Acta

    (2006)
  • M. Allhorn et al.

    Human IgG/Fc gamma R interactions are modulated by streptococcal IgG glycan hydrolysis

    PLoS ONE

    (2008)
  • R.M. Anthony et al.

    A novel role for the IgG Fc glycan: the anti-inflammatory activity of sialylated IgG Fcs

    J. Clin. Immunol.

    (2010)
  • J. Bernard et al.

    Discriminating between protective and enhancing HIV antibodies

    AIDS Res. Hum. Retroviruses

    (1990)
  • P.V. Beum et al.

    Binding of rituximab, trastuzumab, cetuximab, or mAb T101 to cancer cells promotes trogocytosis mediated by THP-1 cells and monocytes

    J. Immunol.

    (2008)
  • R.A. Clynes et al.

    Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets

    Nat. Med.

    (2000)
  • M. Collin et al.

    EndoS and SpeB from Streptococcus pyogenes inhibit immunoglobulin-mediated opsonophagocytosis

    Infect. Immun.

    (2002)
  • S. Dall'Ozzo et al.

    Rituximab-dependent cytotoxicity by natural killer cells: influence of FCGR3A polymorphism on the concentration–effect relationship

    Cancer Res.

    (2004)
  • P.A. Darrah et al.

    Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major

    Nat. Med.

    (2007)
  • H.B. Fleit et al.

    The human monocyte-like cell line THP-1 expresses Fc gamma RI and Fc gamma RII

    J. Leukoc. Biol.

    (1991)
  • B.K. Flesch et al.

    Inhibition of monocyte and polymorphonuclear granulocyte immune phagocytosis by monoclonal antibodies specific for Fc gamma RI, II and III

    Ann. Hematol.

    (1997)
  • Cited by (340)

    View all citing articles on Scopus
    1

    Co-first author.

    View full text