Abstract

Plasma antimalarial Ab can mediate antiparasite immunity but has not previously been characterized at the molecular level. Here, we develop an innovative strategy to characterize humoral responses by integrating profiles of plasma immunoglobulins (IGs) or Abs with those expressed on B cells as part of the B cell receptor. We applied this strategy to define plasma IG and to determine variable (V) gene usage after vaccination with the Plasmodium falciparum zygote antigen Pfs25. Using proteomic tools coupled with bulk immunosequencing data, we determined human antigen-binding fragment [F(ab′)2] peptide sequences from plasma IG of adults who received 4 doses of Pfs25-EPA/Alhydrogel. Specifically, Pfs25 antigen-specific F(ab′)2 peptides (Pfs25-IG) were aligned to cDNA sequences of IG heavy (IGH) chain complementarity determining region 3 from a data set generated by total peripheral B cell immunosequencing of the entire vaccinated population. IGHV4 was the most commonly identified IGHV subgroup of Pfs25-IG, a pattern that was corroborated by V heavy/V light chain sequencing of Pfs25-specific single B cells from 5 vaccinees and by matching plasma Pfs25-IG peptides and V-(D)-J sequences of Pfs25-specific single B cells from the same donor. Among 13 recombinant human mAbs generated from IG sequences of Pfs25-specific single B cells, a single IGHV4 mAb displayed strong neutralizing activity, reducing the number of P. falciparum oocysts in infected mosquitoes by more than 80% at 100 μg/mL. Our approach characterizes the human plasma Ab repertoire in response to the Pfs25-EPA/Alhydrogel vaccine and will be useful for studying circulating Abs in response to other vaccines as well as those induced during infections or autoimmune disorders.

Highlights

  • Despite progress on malaria prevention and treatment [1, 2], eradication of this disease will require novel interventions

  • Purified Pfs25 F(ab′)2 fragments were analyzed by mass spectrometry and mass spectra were matched to sequences in 2 data sets: the international ImMunoGeneTics (IMGT) and the IG heavy (IGH) CDR3 data set (Supplemental Table 2)

  • Matches were more frequent to the public IMGT data set, which contains sequences spanning the entire V region [17], than to the in-house IGH CDR3 data set, which only spans a 43 amino acid (AA) region covering CDR3 (IMGT = 180.6 ± 62.2 peptide matches per subject; IGH CDR3 data set = 56.0 ± 18.9 peptide matches per subject) (Supplemental Figure 3)

Read more

Summary

Introduction

Despite progress on malaria prevention and treatment [1, 2], eradication of this disease will require novel interventions. Transmission blocking vaccines (TBVs) prevent parasite spread through the vector by inducing Abs to surface antigens of mosquito sexual stage development of Plasmodium falciparum [3,4,5,6]. The P. falciparum zygote/ookinete protein Pfs has been the leading TBV candidate antigen for 3 decades and induces Abs that neutralize sexual stage parasites in laboratory assays [7, 8]. Pfs has advanced to clinical trials in endemic settings but has shown limited potency and variable (V) serum functional activity. The molecular definition of the serum Ab repertoire may explain this limitation and guide the design of improved Pfs vaccines.

Methods
Results
Conclusion
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