scholarly journals Daunorubicin-containing CLL1-targeting nanomicelles have anti-leukemia stem cell activity in acute myeloid leukemia

2019 ◽  
Vol 20 ◽  
pp. 102004 ◽  
Author(s):  
Tzu-yin Lin ◽  
Yanjun Zhu ◽  
Yuanpei Li ◽  
Hongyong Zhang ◽  
Ai-Hong Ma ◽  
...  
Oncogene ◽  
2016 ◽  
Vol 36 (18) ◽  
pp. 2589-2598 ◽  
Author(s):  
S Goyama ◽  
M Shrestha ◽  
J Schibler ◽  
L Rosenfeldt ◽  
W Miller ◽  
...  

2021 ◽  
Vol 12 (6) ◽  
Author(s):  
Matthew E. Massett ◽  
Laura Monaghan ◽  
Shaun Patterson ◽  
Niamh Mannion ◽  
Roderick P. Bunschoten ◽  
...  

AbstractEpigenomic dysregulation is a common pathological feature in human hematological malignancies. H3K9me3 emerges as an important epigenomic marker in acute myeloid leukemia (AML). Its associated methyltransferases, such as SETDB1, suppress AML leukemogenesis, whilst H3K9me3 demethylases KDM4C is required for mixed-lineage leukemia rearranged AML. However, the specific role and molecular mechanism of action of another member of the KDM4 family, KDM4A has not previously been clearly defined. In this study, we delineated and functionally validated the epigenomic network regulated by KDM4A. We show that selective loss of KDM4A is sufficient to induce apoptosis in a broad spectrum of human AML cells. This detrimental phenotype results from a global accumulation of H3K9me3 and H3K27me3 at KDM4A targeted genomic loci thereby causing downregulation of a KDM4A-PAF1 controlled transcriptional program essential for leukemogenesis, distinct from that of KDM4C. From this regulatory network, we further extracted a KDM4A-9 gene signature enriched with leukemia stem cell activity; the KDM4A-9 score alone or in combination with the known LSC17 score, effectively stratifies high-risk AML patients. Together, these results establish the essential and unique role of KDM4A for AML self-renewal and survival, supporting further investigation of KDM4A and its targets as a potential therapeutic vulnerability in AML.


2019 ◽  
Vol 76 ◽  
pp. S31
Author(s):  
Christopher Park ◽  
Gaelle Martin ◽  
Nainita Roy ◽  
Sohini Chakraborty ◽  
Alexis Desrichard ◽  
...  

2019 ◽  
Vol 110 (7) ◽  
pp. 2200-2210 ◽  
Author(s):  
Yajing Chu ◽  
Yangpeng Chen ◽  
Mengke Li ◽  
Deyang Shi ◽  
Bichen Wang ◽  
...  

2019 ◽  
Vol 81 ◽  
pp. 27-34
Author(s):  
Bijan Moshaver ◽  
Rolf F. Wouters ◽  
Angèle Kelder ◽  
Gert J. Ossenkoppele ◽  
Guus A.H. Westra ◽  
...  

Oncotarget ◽  
2016 ◽  
Vol 7 (36) ◽  
pp. 57811-57820 ◽  
Author(s):  
Noureldien H.E. Darwish ◽  
Thangirala Sudha ◽  
Kavitha Godugu ◽  
Osama Elbaz ◽  
Hasan A. Abdelghaffar ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2730-2730 ◽  
Author(s):  
Susumu Goyama ◽  
Mahesh Shrestha ◽  
Janet Schibler ◽  
Leah Rosenfeldt ◽  
Whitney Miller ◽  
...  

Abstract Leukemic stem cells (LSCs) are capable of limitless self-renewal and indefinitely propagating leukemia. Eradication of LSCs is the ultimate goal of treating acute myeloid leukemia (AML). Using a mouse model of AML induced by the MLL-fusion protein MLL-AF9, we recently showed that the combined loss of Runx1/Cbfb inhibited the development of leukemia in vivo (Goyama S…Mulloy JC. Transcription factor RUNX1 promotes survival of acute myeloid leukemia cells. Journal of Clinical Investigation 123(9): 3876-3888, 2013). However, LSC-enriched cells with immature surface phenotype (cKit+Gr1-) remained viable in Runx1/Cbfb-deleted MLL-AF9 cells, indicating that RUNX targeting may not eradicate the most immature LSCs. Gene expression analyses of Runx1/Cbfb-deleted MLL-AF9 cells revealed the upregulation of thrombin pathway genes including a thrombin-activatable receptor PAR-1. Interestingly, both overexpression and knockout of PAR-1 inhibit leukemogenesis but do so through distinct mechanisms. Similar to the effect of Runx1/Cbfb-depletion, PAR-1 overexpression induced p21 expression and attenuated proliferation in MLL-AF9 cells. To our surprise, PAR-1-deficiency also prevented leukemia development induced by a small number of MLL-AF9 LSCs in vivo. Re-expression of PAR-1 in PAR-1-deficient cells combined with a limiting-dilution transplantation assay demonstrated the cell-dose dependent role of PAR-1 in MLL-AF9 leukemia: PAR-1 inhibited rapid leukemic proliferation when there are a large number of LSCs, while a small numbers of LSCs required PAR-1 for their growth. Mechanistically, PAR-1 increased adhering properties of MLL-AF9 cells and promoted their engraftment to bone marrow. PAR-1-deficiency also reduced leukemogenicity of AML1-ETO-induced leukemia. Together, these data reveal a multifaceted role for PAR-1 in leukemogenesis, and highlight this receptor as a potential target to eradicate primitive LSCs in AML. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document