Developmental differences in megakaryocytopoiesis are associated with up-regulated TPO signaling through mTOR and elevated GATA-1 levels in neonatal …

ZJ Liu, J Italiano Jr, F Ferrer-Marin… - Blood, The Journal …, 2011 - ashpublications.org
ZJ Liu, J Italiano Jr, F Ferrer-Marin, R Gutti, M Bailey, B Poterjoy, L Rimsza, M Sola-Visner
Blood, The Journal of the American Society of Hematology, 2011ashpublications.org
Multiple observations support the existence of developmental differences in
megakaryocytopoiesis. We have previously shown that neonatal megakaryocyte (MK)
progenitors are hyperproliferative and give rise to MKs smaller and of lower ploidy than adult
MKs. Based on these characteristics, neonatal MKs have been considered immature. The
molecular mechanisms underlying these differences are unclear, but contribute to the
pathogenesis of disorders of neonatal megakaryocytopoiesis. In the present study, we …
Abstract
Multiple observations support the existence of developmental differences in megakaryocytopoiesis. We have previously shown that neonatal megakaryocyte (MK) progenitors are hyperproliferative and give rise to MKs smaller and of lower ploidy than adult MKs. Based on these characteristics, neonatal MKs have been considered immature. The molecular mechanisms underlying these differences are unclear, but contribute to the pathogenesis of disorders of neonatal megakaryocytopoiesis. In the present study, we demonstrate that low-ploidy neonatal MKs, contrary to traditional belief, are more mature than adult low-ploidy MKs. These mature MKs are generated at a 10-fold higher rate than adult MKs, and result from a developmental uncoupling of proliferation, polyploidization, and terminal differentiation. This pattern is associated with up-regulated thrombopoietin (TPO) signaling through mammalian target of rapamycin (mTOR) and elevated levels of full-length GATA-1 and its targets. Blocking of mTOR with rapamycin suppressed the maturation of neonatal MKs without affecting ploidy, in contrast to the synchronous inhibition of polyploidization and cytoplasmic maturation in adult MKs. We propose that these mechanisms allow fetuses/neonates to populate their rapidly expanding bone marrow and intravascular spaces while maintaining normal platelet counts, but also set the stage for disorders restricted to fetal/neonatal MK progenitors, including the Down syndrome–transient myeloproliferative disorder and the thrombocytopenia absent radius syndrome.
ashpublications.org