Inflammatory signaling regulates embryonic hematopoietic stem and progenitor cell production

  1. Nancy A. Speck1,2,10
  1. 1Abramson Family Cancer Research Institute,
  2. 2Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19014, USA;
  3. 3Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02115, USA;
  4. 4Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA;
  5. 5Howard Hughes Medical Institute, Division of Hematology/Oncology, Boston Children’s Hospital, Boston, Massachusetts 02115, USA;
  6. 6The University of Edinburgh, Edinburgh EH8 9YL, United Kingdom;
  7. 7Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA;
  8. 8Department of Bioengineering, University of Iowa, Iowa City, Iowa 52242, USA
  1. Corresponding authors: nancyas{at}exchange.upenn.edu, tnorth{at}bidmc.harvard.edu
  1. 10 These authors contributed equally to this work.

  • 9 Present address: Children’s Research Institute, University of Texas Southwestern Medical School, Dallas, Texas 75235, USA.

Abstract

Identifying signaling pathways that regulate hematopoietic stem and progenitor cell (HSPC) formation in the embryo will guide efforts to produce and expand HSPCs ex vivo. Here we show that sterile tonic inflammatory signaling regulates embryonic HSPC formation. Expression profiling of progenitors with lymphoid potential and hematopoietic stem cells (HSCs) from aorta/gonad/mesonephros (AGM) regions of midgestation mouse embryos revealed a robust innate immune/inflammatory signature. Mouse embryos lacking interferon γ (IFN-γ) or IFN-α signaling and zebrafish morphants lacking IFN-γ and IFN-ϕ activity had significantly fewer AGM HSPCs. Conversely, knockdown of IFN regulatory factor 2 (IRF2), a negative regulator of IFN signaling, increased expression of IFN target genes and HSPC production in zebrafish. Chromatin immunoprecipitation (ChIP) combined with sequencing (ChIP-seq) and expression analyses demonstrated that IRF2-occupied genes identified in human fetal liver CD34+ HSPCs are actively transcribed in human and mouse HSPCs. Furthermore, we demonstrate that the primitive myeloid population contributes to the local inflammatory response to impact the scale of HSPC production in the AGM region. Thus, sterile inflammatory signaling is an evolutionarily conserved pathway regulating the production of HSPCs during embryonic development.

Keywords

Footnotes

  • Received September 28, 2014.
  • Accepted October 29, 2014.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance