[PDF][PDF] Single-cell analysis of neonatal HSC ontogeny reveals gradual and uncoordinated transcriptional reprogramming that begins before birth

Y Li, W Kong, W Yang, RM Patel, EB Casey… - Cell stem cell, 2020 - cell.com
Y Li, W Kong, W Yang, RM Patel, EB Casey, T Okeyo-Owuor, JM White, SN Porter, SA Morris
Cell stem cell, 2020cell.com
Fetal and adult hematopoietic stem cells (HSCs) have distinct proliferation rates, lineage
biases, gene expression profiles, and gene dependencies. Although these differences are
widely recognized, it is not clear how the transition from fetal to adult identity is coordinated.
Here we show that murine HSCs and committed hematopoietic progenitor cells (HPCs)
undergo a gradual, rather than precipitous, transition from fetal to adult transcriptional states.
The transition begins prior to birth and is punctuated by a late prenatal spike in type I …
Summary
Fetal and adult hematopoietic stem cells (HSCs) have distinct proliferation rates, lineage biases, gene expression profiles, and gene dependencies. Although these differences are widely recognized, it is not clear how the transition from fetal to adult identity is coordinated. Here we show that murine HSCs and committed hematopoietic progenitor cells (HPCs) undergo a gradual, rather than precipitous, transition from fetal to adult transcriptional states. The transition begins prior to birth and is punctuated by a late prenatal spike in type I interferon signaling that promotes perinatal HPC expansion and sensitizes progenitors to the leukemogenic FLT3ITD mutation. Most other changes in gene expression and enhancer activation are imprecisely timed and poorly coordinated. Thus, heterochronic enhancer elements, and their associated transcripts, are activated independently of one another rather than as part of a robust network. This simplifies the regulatory programs that guide neonatal HSC/HPC ontogeny, but it creates heterogeneity within these populations.
cell.com