4.7 Article

RUNX1-deficient human megakaryocytes demonstrate thrombopoietic and platelet half-life and functional defects

Journal

BLOOD
Volume 141, Issue 3, Pages 260-270

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2022017561

Keywords

-

Categories

Ask authors/readers for more resources

Heterozygous defects in RUNX1 are responsible for a familial platelet disorder with associated myeloid malignancy (FPDMM). An in vitro megakaryopoiesis system was developed to mimic the FPDMM quantitative megakaryocyte defect by decreasing RUNX1 expression. The shRX-megakaryocytes released fewer platelets, had a diminished half-life, and showed poor responsiveness to agonists. A small-molecule inhibitor RepSox corrected the thrombopoietic defect and improved the defects in thrombus formation and platelet half-life.
Heterozygous defects in runt-related transcription factor 1 (RUNX1) are causative of a familial platelet disorder with associated myeloid malignancy (FPDMM). Because RUNX1-deficient animal models do not mimic bleeding disorder or leukemic risk associated with FPDMM, development of a proper model system is critical to understanding the underlying mechanisms of the observed phenotype and to identifying therapeutic interventions. We previously reported an in vitro megakaryopoiesis system comprising human CD34(+) hematopoietic stem and progenitor cells that recapitulated the FPDMM quantitative megakaryocyte defect through a decrease in RUNX1 expression via a lentiviral short hairpin RNA strategy. We now show that shRX-megakaryocytes have a marked reduction in agonist responsiveness. We then infused shRX-megakaryocytes into immunocompromised NOD scid gamma (NSG) mice and demonstrated that these megakaryocytes released fewer platelets than megakaryocytes transfected with a nontargeting shRNA, and these platelets had a diminished half-life. The platelets were also poorly responsive to agonists, unable to correct thrombus formation in NSG mice homozygous for a R1326H mutation in von Willebrand Factor (VWFR1326H), which switches the species-binding specificity of the VWF from mouse to human glycoprotein Ib alpha. A small-molecule inhibitor RepSox, which blocks the transforming growth factor beta 1 (TGF beta 1) pathway and rescued defective megakaryopoiesis in vitro, corrected the thrombopoietic defect, defects in thrombus formation and platelet half-life, and agonist response in NSG/VWFR1326H mice. Thus, this model recapitulates the defects in FPDMM megakaryocytes and platelets, identifies previously unrecognized defects in thrombopoiesis and platelet half-life, and demonstrates for the first time, reversal of RUNX1 deficiency-induced hemostatic defects by a drug.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available