A unified model of human hemoglobin switching through single-cell genome editing

Nat Commun. 2021 Aug 17;12(1):4991. doi: 10.1038/s41467-021-25298-9.

Abstract

Key mechanisms of fetal hemoglobin (HbF) regulation and switching have been elucidated through studies of human genetic variation, including mutations in the HBG1/2 promoters, deletions in the β-globin locus, and variation impacting BCL11A. While this has led to substantial insights, there has not been a unified understanding of how these distinct genetically-nominated elements, as well as other key transcription factors such as ZBTB7A, collectively interact to regulate HbF. A key limitation has been the inability to model specific genetic changes in primary isogenic human hematopoietic cells to uncover how each of these act individually and in aggregate. Here, we describe a single-cell genome editing functional assay that enables specific mutations to be recapitulated individually and in combination, providing insights into how multiple mutation-harboring functional elements collectively contribute to HbF expression. In conjunction with quantitative modeling and chromatin capture analyses, we illustrate how these genetic findings enable a comprehensive understanding of how distinct regulatory mechanisms can synergistically modulate HbF expression.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • CRISPR-Cas Systems
  • Chromatin
  • Chromosomes
  • DNA-Binding Proteins / metabolism
  • Fetal Hemoglobin / genetics
  • Fetal Hemoglobin / metabolism
  • Gene Editing*
  • Gene Expression
  • Globins
  • Hemoglobins / genetics*
  • Hemoglobins / metabolism*
  • Humans
  • Mutation
  • Repressor Proteins
  • Transcription Factors / metabolism
  • beta-Globins / genetics

Substances

  • BCL11A protein, human
  • Chromatin
  • DNA-Binding Proteins
  • Hemoglobins
  • Repressor Proteins
  • Transcription Factors
  • ZBTB7A protein, human
  • beta-Globins
  • Globins
  • Fetal Hemoglobin