Emerging Microbes and Infections (Dec 2024)

Deep immunoglobulin repertoire sequencing depicts a comprehensive atlas of spike-specific antibody lineages shared among COVID-19 convalescents

  • Qihong Yan,
  • Yudi Zhang,
  • Ruitian Hou,
  • Wenjing Pan,
  • Huan Liang,
  • Xijie Gao,
  • Weiqi Deng,
  • Xiaohan Huang,
  • Linbing Qu,
  • Congli Tang,
  • Ping He,
  • Banghui Liu,
  • Qian Wang,
  • Xinwei Zhao,
  • Zihan Lin,
  • Zhaoming Chen,
  • Pingchao Li,
  • Jian Han,
  • Xiaoli Xiong,
  • Jincun Zhao,
  • Song Li,
  • Xuefeng Niu,
  • Ling Chen

DOI
https://doi.org/10.1080/22221751.2023.2290841
Journal volume & issue
Vol. 13, no. 1

Abstract

Read online

ABSTRACTNeutralizing antibodies are a key component in protective humoral immunity against SARS-CoV-2. Currently, available technologies cannot track epitope-specific antibodies in global antibody repertoires. Thus, the comprehensive repertoire of spike-specific neutralizing antibodies elicited by SARS-CoV-2 infection is not fully understood. We therefore combined high-throughput immunoglobulin heavy chain (IgH) repertoire sequencing, and structural and bioinformatics analysis to establish an antibodyomics pipeline, which enables tracking spike-specific antibody lineages that target certain neutralizing epitopes. We mapped the neutralizing epitopes on the spike and determined the epitope-preferential antibody lineages. This analysis also revealed numerous overlaps between immunodominant neutralizing antibody-binding sites and mutation hotspots on spikes as observed so far in SARS-CoV-2 variants. By clustering 2677 spike-specific antibodies with 360 million IgH sequences that we sequenced, a total of 329 shared spike-specific antibody clonotypes were identified from 33 COVID-19 convalescents and 24 SARS-CoV-2-naïve individuals. Epitope mapping showed that the shared antibody responses target not only neutralizing epitopes on RBD and NTD but also non-neutralizing epitopes on S2. The immunodominance of neutralizing antibody response is determined by the occurrence of specific precursors in human naïve B-cell repertoires. We identified that only 28 out of the 329 shared spike-specific antibody clonotypes persisted for at least 12 months. Among them, long-lived IGHV3-53 antibodies are likely to evolve cross-reactivity to Omicron variants through accumulating somatic hypermutations. Altogether, we created a comprehensive atlas of spike-targeting antibody lineages in COVID-19 convalescents and antibody precursors in human naïve B cell repertoires, providing a valuable reference for future vaccine design and evaluation.

Keywords