Sustained fetal hematopoiesis causes juvenile death from leukemia: evidence from a dual-age–specific mouse model

N Vara, Y Liu, Y Yan, SY Lensing, N Colorado… - Blood …, 2020 - ashpublications.org
N Vara, Y Liu, Y Yan, SY Lensing, N Colorado, D Robinson, J Zhang, X Zhang, EA Peterson
Blood Advances, 2020ashpublications.org
It is not clear whether disrupted age-specific hematopoiesis contributes to the complex
manifestations in leukemia patients who carry identical mutations, particularly in pediatric
and adult patients with similar clinical characteristics. By studying a dual-age–specific
mouse model, we demonstrate that (1) loss of Pten during the fetal-to-adult hematopoiesis
switch (hematopoiesis switch) causes sustained fetal hematopoiesis, resulting in death in
juvenile leukemia;(2) myeloid-biased hematopoiesis in juvenile mice is associated with the …
Abstract
It is not clear whether disrupted age-specific hematopoiesis contributes to the complex manifestations in leukemia patients who carry identical mutations, particularly in pediatric and adult patients with similar clinical characteristics. By studying a dual-age–specific mouse model, we demonstrate that (1) loss of Pten during the fetal-to-adult hematopoiesis switch (hematopoiesis switch) causes sustained fetal hematopoiesis, resulting in death in juvenile leukemia; (2) myeloid-biased hematopoiesis in juvenile mice is associated with the sustained fetal properties of hematopoietic stem cells (HSCs); (3) the age specificity of juvenile myelomonocytic leukemia depends on the copy number of Pten and Nf1; (4) single-allelic Pten deletion during the hematopoiesis switch causes constitutive activation of MAPK in juvenile mice with Nf1 loss of heterozygosity (LOH); and (5) Nf1 LOH causes monocytosis in juvenile mice with Pten haploinsufficiency but does not cause lethality until adulthood. Our data suggest that 1 copy of Pten is sufficient to maintain an intact negative-feedback loop of the Akt pathway and HSC function in reconstitution, despite MAPK being constitutively activated in juvenile Pten+/ΔNf1LOH mice. However, 2 copies of Pten are required to maintain the integrity of the MAPK pathway in juvenile mice with Nf1 haploinsufficiency. Our data indicate that previous investigations of Pten function in wild-type mice may not reflect the impact of Pten loss in mice with Nf1 mutations or other genetic defects. We provide a proof of concept that disassociated age-specific hematopoiesis contributes to leukemogenesis and pediatric demise.
ashpublications.org