scholarly journals Maternal exposure to fine particulate matter during pregnancy induces progressive senescence of hematopoietic stem cells under preferential impairment of the bone marrow microenvironment and aids development of myeloproliferative disease

Leukemia ◽  
2019 ◽  
Vol 34 (5) ◽  
pp. 1481-1484 ◽  
Author(s):  
Govinda Bhattarai ◽  
Jae Bong Lee ◽  
Min-Hye Kim ◽  
Suhan Ham ◽  
Han-Sol So ◽  
...  
Cell ◽  
2007 ◽  
Vol 129 (6) ◽  
pp. 1081-1095 ◽  
Author(s):  
Carl R. Walkley ◽  
Jeremy M. Shea ◽  
Natalie A. Sims ◽  
Louise E. Purton ◽  
Stuart H. Orkin

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1356-1356
Author(s):  
Christian Brandts ◽  
Miriam Rode ◽  
Beate Lindtner ◽  
Gabriele Koehler ◽  
Steffen Koschmieder ◽  
...  

Abstract Activating mutations in Flt3, N- and K-Ras have been reported in all AML subtypes and represent common molecular defects in de novo AML. We have previously shown that these mutations lead to constitutive AKT phosphorylation and activation. As a consequence, Akt phosphorylation is found in myeloid blasts of the majority of AML patients. We reasoned that constitutively active AKT may contribute to leukemia development, and therefore we assessed the contribution of AKT in oncogenic transformation in vivo. For this purpose, we established an inducible mouse model expressing myristylated AKT1 under the control of the scl-3′ enhancer (MyrAKT1). This system restricts activated AKT1 to endothelium, hematopoietic stem cells and myeloid lineage cells at a low but detectable level. About 40% of induced mice developed a myeloproliferative disorder after latencies of 7 to 22 months. Onset of disease was frequently associated with hemangioma formation, due to endothelial MyrAKT1 expression. The myeloproliferative disorder was associated with splenomegaly with increased extramedullary hematopoiesis, while the peripheral blood contained mature granulocytes. Furthermore, the stem cell and progenitor cell compartment in spleens and bone marrow of these mice was altered compared to control mice. Colony formation assays with MyrAKT1-expressing bone marrow suggested that overactivation of AKT1 enhanced proliferation. The AKT1-induced disease was transplantable by both bone marrow and spleen cells. These findings highlight the oncogenic capacity of constitutively activated AKT1 in vivo and indicate that AKT is an attractive target for therapeutic intervention in AML.


Blood ◽  
2010 ◽  
Vol 115 (7) ◽  
pp. 1406-1415 ◽  
Author(s):  
Michael G. Kharas ◽  
Rachel Okabe ◽  
Jared J. Ganis ◽  
Maricel Gozo ◽  
Tulasi Khandan ◽  
...  

Abstract Human cancers, including acute myeloid leukemia (AML), commonly display constitutive phosphoinositide 3-kinase (PI3K) AKT signaling. However, the exact role of AKT activation in leukemia and its effects on hematopoietic stem cells (HSCs) are poorly understood. Several members of the PI3K pathway, phosphatase and tensin homolog (Pten), the forkhead box, subgroup O (FOXO) transcription factors, and TSC1, have demonstrated functions in normal and leukemic stem cells but are rarely mutated in leukemia. We developed an activated allele of AKT1 that models increased signaling in normal and leukemic stem cells. In our murine bone marrow transplantation model using a myristoylated AKT1 (myr-AKT), recipients develop myeloproliferative disease, T-cell lymphoma, or AML. Analysis of the HSCs in myr-AKT mice reveals transient expansion and increased cycling, associated with impaired engraftment. myr-AKT–expressing bone marrow cells are unable to form cobblestones in long-term cocultures. Rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR) rescues cobblestone formation in myr-AKT–expressing bone marrow cells and increases the survival of myr-AKT mice. This study demonstrates that enhanced AKT activation is an important mechanism of transformation in AML and that HSCs are highly sensitive to excess AKT/mTOR signaling.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1420-1420
Author(s):  
S. Haihua Chu ◽  
Diane Heiser ◽  
Li Li ◽  
Ian M Kaplan ◽  
Curt I. Civin ◽  
...  

Abstract Abstract 1420 Poster Board I-443 As one of the most common genetic alterations found in acute myeloid leukemia (AML), constitutive activation of the FMS-like tyrosine kinase (FLT3) has provided a promising candidate for small molecule targeted therapy. However, the results of FLT3 inhibitor monotherapy trials indicate FLT3 inhibition alone is insufficient to induce consistent and durable responses. Moreover, after an initial response, many patients relapse, suggesting that leukemia-initiating stem cells may be escaping inhibitor-induced cytotoxicity. Currently, the exact stage at which activating mutations in FLT3 occur during transformation is unknown. While FLT3 knockout mice have minor defects in hematopoiesis, very little is known about either the effect of FLT3 activating mutations on normal hematopoietic stem cells or the contribution of FLT3 activation to leukemogenesis. Thus, in order to better understand the underlying molecular mechanisms of transformation and to identify novel targets for treatment of AML, the role of FLT3 activating mutations in hematopoietic stem cells (HSCs) is of great interest. To study the natural stem cell reservoir and other populations that may escape inhibition, our laboratory has developed a knock-in mouse model in which the FLT3/ITD mutation (an internal tandem duplication correlated with poor prognosis in patients) has been introduced under the endogenous promoter, resulting in myeloproliferative disease (MPD). Conventional transplantation using unfractionated or lineage-depleted marrow from FLT3/ITD mice failed to fully engraft or recapitulate disease, suggesting a HSC defect. Thus, in order to identify a compartment enriched for MPD-initiating cells, several cell surface marker-defined hematopoietic populations were transplanted and compared for engraftment and disease recapitulation. Lineage negative (LIN-), KSL (KIT+SCA+LIN-), MPP (KSL CD34+FLT3+), and ST-HSC (KSL CD34+FLT3-) cells sorted from FLT3/ITD bone marrow all had significantly reduced reconstitution capacity compared to the same compartment from WT littermates. In contrast, highly purified LT-HSCs (KSL CD34-FLT3-) generated equivalent engraftment whether from WT or ITD bone marrow. Furthermore, we measured Hoechst dye efflux in WT and ITD bone marrow to examine side population (SP) cells, known to be enriched in HSC activity, and found that FLT3/ITD mice displayed five-fold fewer SP cells. In addition, bone marrow from FLT3/ITD mice showed a ten-fold decrease in SLAM-defined stem cell frequency (LIN-CD48-CD41-CD150+). 500 sorted SLAM cells from either WT or FLT3/ITD mice were sufficient to fully reconstitute a transplant recipient, demonstrating an equivalent engraftment capacity within this HSC-enriched compartment. Moreover, the MPD phenotype was successfully recapitulated in primary transplants of FLT3/ITD SLAM cells as characterized by an increase in myeloid progenitors, expansion of the LIN- fraction, enlarged spleens and depletion of the SLAM compartment compared to WT SLAM transplant recipients. Classically defined as a class II oncogene, FLT3 activating mutations have been shown to drive proliferation in cells harboring the mutation. To investigate whether FLT3/ITD drives proliferation in the most primitive hematopoietic compartments, BrdU incorporation was examined in FLT3/ITD hematopoietic stem and progenitor-enriched subsets. While myeloid progenitor compartments showed a decrease in proliferation as compared to WT littermates, the FLT3/ITD SLAM and KSL compartments had an increased percentage of BrdU-incorporating cells. Altogether, our data suggests a role for FLT3/ITD in driving normally quiescent HSCs to proliferate, thereby depleting the pool of primitive HSCs. In this model, HSC depletion coupled to rapid expansion in progenitor cell numbers leads to perturbation of normal hematopoiesis giving rise to a myeloproliferative disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document