scholarly journals Canonical Notch Signaling Is Dispensable for the Maintenance of Adult Hematopoietic Stem Cells

2008 ◽  
Vol 2 (4) ◽  
pp. 356-366 ◽  
Author(s):  
Ivan Maillard ◽  
Ute Koch ◽  
Alexis Dumortier ◽  
Olga Shestova ◽  
Lanwei Xu ◽  
...  
2017 ◽  
Vol 51 ◽  
pp. 1-6.e2 ◽  
Author(s):  
Qiuping He ◽  
Suwei Gao ◽  
Junhua Lv ◽  
Wei Li ◽  
Feng Liu

2008 ◽  
Vol 3 (3) ◽  
pp. 314-326 ◽  
Author(s):  
Thomas Mercher ◽  
Melanie G. Cornejo ◽  
Christopher Sears ◽  
Thomas Kindler ◽  
Sandra A. Moore ◽  
...  

2005 ◽  
Vol 82 (4) ◽  
pp. 285-294 ◽  
Author(s):  
Takahiro Suzuki ◽  
Shigeru Chiba

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 267-267 ◽  
Author(s):  
Ivan Maillard ◽  
Seth E. Pross ◽  
Olga Shestova ◽  
Hong Sai ◽  
Jon C. Aster ◽  
...  

Abstract Canonical Notch signaling operates through a highly conserved pathway that regulates the differentiation and homeostasis of hematopoietic cells. Ligand-receptor binding initiates proteolytic release of the Notch intracellular domain (ICN) which migrates to the nucleus, binds the transcription factor CSL/RBPJk and activates target genes through the recruitment of transcriptional coactivators of the Mastermind-like family (MAML). Notch signaling is essential for the emergence of hematopoietic stem cells (HSCs) during fetal life, but its effects on adult HSCs are controversial. In gain-of-function experiments, activation of Notch signaling in adult HSCs increased their self-renewal potential in vitro and in vivo. However, loss-of-function studies have provided conflicting results as to the role of physiological Notch signaling in HSC maintenance and homeostasis. To address this question, we expressed DNMAML1, a GFP-tagged pan-inhibitor of Notch signaling, in mouse HSCs. We have shown previously that DNMAML1 interferes with the formation of the ICN/CSL/MAML transcriptional activation complex and blocks signaling from all four Notch receptors (Notch1-4) (Maillard, Blood 2004). Transfer of DNMAML1-transduced bone marrow (BM) as compared to control GFP-transduced BM into lethally irradiated recipients gave rise to similar long-term stable expression of GFP for at least 6 months after transplant. DNMAML1 and GFP-transduced cells contributed equally to all hematopoietic lineages, except to the T cell and marginal zone B cell lineages, which are Notch-dependent. Expression of DNMAML1 did not affect the size of the BM progenitor compartment (Lin negative, Sca-1 positive, c-Kit high, or LSK cells), or the proportion of LSK cells that were negative for Flt3 and L-Selectin expression (containing long-term HSCs). The stem cell function of DNMAML1-transduced LSK cells was further assessed with in vivo competitive repopulation assays in lethally irradiated recipients. DNMAML1 and GFP-transduced LSK cells competed equally well with wild-type BM, as judged by their contribution to the myeloid lineage up to 4 months post-transplant, through two successive rounds of transplantation. Our data indicate that canonical Notch signaling is dispensable for the maintenance of stem cell function in adult HSCs.


2014 ◽  
Vol 33 (20) ◽  
pp. 2363-2373 ◽  
Author(s):  
Albert D Kim ◽  
Chase H Melick ◽  
Wilson K Clements ◽  
David L Stachura ◽  
Martin Distel ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 395-395 ◽  
Author(s):  
Mark Y Chiang ◽  
Olga Shestova ◽  
M. Eden Childs ◽  
Candice Romany ◽  
Jon Aster ◽  
...  

Abstract Abstract 395 The Leukemia Stem Cell (LSC) hypothesis proposes that a subset of the cells in the bulk tumor cell population is responsible for leukemia maintenance and propagation. We tested the LSC hypothesis in murine T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) induced by retroviruses expressing strong gain-of-function Notch1 alleles. This model is relevant to human disease, as Notch1 is activated in ∼70% of human T-ALLs. By transferring sorted leukemia cells to syngeneic secondary recipients at limiting dilution, we found that LSC activity was enriched in the CD8+CD4-HSAhi (Immature Single Positive or “ISP”) T-cell subset. The LSC frequency was ∼1 in 1000 cells, two orders of magnitude higher than in the CD8+CD4+ (Double positive or “DP”) T-cell subset. We found similar results in a KrasG12D-driven T-ALL mouse model where activating Notch mutations occur spontaneously in ∼80% cases. Surprisingly, we were unable to isolate Notch-activated hematopoietic stem and progenitor cells (HSPCs) to test for LSC activity. Upon further analysis, we observed that activation of Notch in HSPCs caused loss of stem cell quiescence and progressive, complete loss of long-term hematopoietic stem cells (LT-HSCs) over several weeks. Notch signals had no significant effects on stem cell homing, apoptosis, or senescence. Similar results were obtained in both noncompetitive and competitive secondary transplants as well as in a mouse model in which Notch activation is conditionally induced in HSCs by Cre recombinase. We conclude that while Notch signaling promotes LSC activity, it extinguishes HSCs. These results provide a rationale for therapeutic targeting of the ISP-like T-cell subset in Notch-activated T-ALL while underscoring the potential difficulty of manipulating Notch signaling to expand normal, long-term stem cell populations for clinical applications. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document