PLoS Genetics (Jan 2013)

Growth factor independent-1 maintains Notch1-dependent transcriptional programming of lymphoid precursors.

  • James D Phelan,
  • Ingrid Saba,
  • Hui Zeng,
  • Christian Kosan,
  • Malynda S Messer,
  • H Andre Olsson,
  • Jennifer Fraszczak,
  • David A Hildeman,
  • Bruce J Aronow,
  • Tarik Möröy,
  • H Leighton Grimes

DOI
https://doi.org/10.1371/journal.pgen.1003713
Journal volume & issue
Vol. 9, no. 9
p. e1003713

Abstract

Read online

Growth factor independent 1 (Gfi1) is a transcriptional repressor originally identified as a gene activated in T-cell leukemias induced by Moloney-murine-leukemia virus infection. Notch1 is a transmembrane receptor that is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL). Gfi1 is an important factor in the initiation and maintenance of lymphoid leukemias and its deficiency significantly impedes Notch dependent initiation of T-ALL in animal models. Here, we show that immature hematopoietic cells require Gfi1 to competently integrate Notch-activated signaling. Notch1 activation coupled with Gfi1 deficiency early in T-lineage specification leads to a dramatic loss of T-cells, whereas activation in later stages leaves development unaffected. In Gfi1 deficient multipotent precursors, Notch activation induces lethality and is cell autonomous. Further, without Gfi1, multipotent progenitors do not maintain Notch1-activated global expression profiles typical for T-lineage precursors. In agreement with this, we find that both lymphoid-primed multipotent progenitors (LMPP) and early T lineage progenitors (ETP) do not properly form or function in Gfi1(-/-) mice. These defects correlate with an inability of Gfi1(-/-) progenitors to activate lymphoid genes, including IL7R, Rag1, Flt3 and Notch1. Our data indicate that Gfi1 is required for hematopoietic precursors to withstand Notch1 activation and to maintain Notch1 dependent transcriptional programming to determine early T-lymphoid lineage identity.