Abstract

Antibody-secreting cells (ASC), plasmablasts and plasma cells, are terminally differentiated B cells responsible for large-scale production and secretion of antibodies. ASC are derived from activated B cells, which may differentiate extrafollicularly or form germinal center (GC) reactions within secondary lymphoid organs. ASC therefore consist of short-lived, poorly matured plasmablasts that generally secrete lower-affinity antibodies, or long-lived, highly matured plasma cells that generally secrete higher-affinity antibodies. The ASC population is responsible for producing an immediate humoral B cell response, the polyclonal antibody repertoire, as well as in parallel building effective humoral memory and immunity, or potentially driving pathology in the case of autoimmunity. ASC are phenotypically and transcriptionally distinct from other B cells and further distinguishable by morphology, varied lifespans, and anatomical localization. Single cell analyses are required to interrogate the functional and transcriptional diversity of ASC and their secreted antibody repertoire and understand the contribution of individual ASC responses to the polyclonal humoral response. Here we summarize the current and emerging functional and molecular techniques for high-throughput characterization of ASC with single cell resolution, including flow and mass cytometry, spot-based and microfluidic-based assays, focusing on functional approaches of the secreted antibodies: specificity, affinity, and secretion rate.

Highlights

  • Antibody-secreting cells (ASC) are B cells that have differentiated following activation to secrete various soluble isotypes of their immunoglobulin receptor with the purpose of binding their target antigen throughout the body [1]

  • We reported in Gerard et al [69] the CelliGo assay using a double fluorescent sandwich ELISA in microfluidic droplets for the identification, sorting, and VH-VL sequencing of antigen-specific IgG antibodies produced by ASC from immunized mice

  • We demonstrated screening of a bacterial antigen, an autoantigen linked to Rheumatoid Arthritis (Glucose-6-phosphate Isomerase), and an insoluble, membrane-expressed antigen; antigen-specific ASC against all 3 model antigens were able to be sorted by this flowed droplet microfluidic technique

Read more

Summary

INTRODUCTION

Antibody-secreting cells (ASC) are B cells that have differentiated following activation to secrete various soluble isotypes of their immunoglobulin receptor with the purpose of binding their target antigen throughout the body [1]. Plasma cells are a long-lived ASC subset, characterized by reinforced expression of genes within the ASC differentiation network, responsible for secreting large quantities of antibodies from within their survival niches. ASC contribute to both the acute humoral response to infection by rapidly generating early antibodies at sites of infection as well as later secreting higher affinity antibodies produced by germinal center reactions to aid in pathogen clearance and protective immunity. While the ASC response is advantageous during infection and when co-opted for immunization, emergence of ASC secreting antibodies towards self-antigens is a deleterious factor in many autoimmune disorders [10]. Despite their importance, much is still unknown regarding ASC differentiation, selection, and heterogeneity, in autoimmune disorders. This review will outline current and emerging HT techniques to characterize single ASC (Figure 1), with discussion of recent applications of these techniques to study the role of ASC in various pathologies as well as to expand understanding of fundamental ASC biology

FUNCTIONAL ANALYSES
MOLECULAR ANALYSES
Multiplexing Is the Future for High Throughput Single ASC Analyses
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call