Allogeneic hematopoietic stem cell (HSC) transplantation is the gold standard therapy for numerous hematological disorders, but limitations exist in HSC number available per donor and our relative inability to successfully expand transplantable HSCs in vitro. In recent years, promising new molecules have been discovered, which are able to expand HSCs in vitro and improve transplantation in vivo in immunodeficient mouse models and non-human primates (e.g. SR1 and UM171).

Here we demonstrate that acute activation of the receptor tyrosine kinase, RET, by its key ligand and co-receptor (GDNF/GFRα1), significantly improves the outgrowth of HSCs invitro and long-term engraftment in immunodeficient Kit-mutant (KitW41/W41) mice.

The expression of RET at the cell surface of cord blood-derived hematopoietic stem and progenitor cells (HSPCs, CD34+CD38-REThi) enriches for stem cell frequency in limiting dilution transplantation assays (~1 in 135), with CD34+CD38-RETlow cells showing low stem cell frequency with limited multilineage engraftment (~1 in 531). Activation of RET in vitro with a single dose of GDNF/GFRα1 reduces apoptosis at early time points, leading to increased HSC expansion over a 7-day period (>4-fold increase vs control). Transplantation of GDNF/GFRα1 treated HSPCs significantly improves engraftment of primary mice, comparable to the previously published SR1/UM171 treatment, and combination treatment of GDNF/GFRα1 with SR1/UM171 improves engraftment compared to individual GDNF/GFRα1 or SR1/UM171 treatments alone.

RET is a single pass transmembrane receptor tyrosine kinase, governing a variety of downstream signalling pathways. Interrogation of the dynamic kinome changes governed by RET in HSPCs, using PAMGene kinome array kinetic technology and single cell mass cytometry revealed key phosphorylation cascades responsive to GDNF/GFRα1 treatment. Key cascades including anti-apoptotic p53 phosphorylation, NFκB activation and increased CD44 cell surface expression, revealed important cellular programs for HSC survival, outgrowth and engraftment activated by GDNF/GFRα1.

Our results provide a promising new target to improve the outgrowth of transplantable HSCs for clinical use, with multiple downstream functions activated for successful transplantation.

Disclosures

No relevant conflicts of interest to declare.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution