Please use this identifier to cite or link to this item: https://doi.org/10.1182/blood-2013-07-511907
DC FieldValue
dc.titlePhysiologic hypoxia promotes maintenance of CML stem cells despite effective BCR-ABL1 inhibition
dc.contributor.authorNg K.P.
dc.contributor.authorManjeri A.
dc.contributor.authorLee K.L.
dc.contributor.authorHuang W.
dc.contributor.authorTan S.Y.
dc.contributor.authorChuah C.T.H.
dc.contributor.authorPoellinger L.
dc.contributor.authorOng S.T.
dc.date.accessioned2020-02-03T09:23:48Z
dc.date.available2020-02-03T09:23:48Z
dc.date.issued2014
dc.identifier.citationNg K.P., Manjeri A., Lee K.L., Huang W., Tan S.Y., Chuah C.T.H., Poellinger L., Ong S.T. (2014). Physiologic hypoxia promotes maintenance of CML stem cells despite effective BCR-ABL1 inhibition. Blood 123 (21) : 3316-3326. ScholarBank@NUS Repository. https://doi.org/10.1182/blood-2013-07-511907
dc.identifier.issn64971
dc.identifier.urihttps://scholarbank.nus.edu.sg/handle/10635/164266
dc.description.abstractC-abl oncogene 1, nonreceptor tyrosine kinase (ABL1) kinase inhibitors such as imatinib mesylate (imatinib) are effective in managing chronic myeloid leukemia (CML) but incapable of eliminating leukemia stem cells (LSCs), suggesting that kinase-independent pathways support LSC survival. Given that the bone marrow (BM) hypoxic microenvironment supports hematopoietic stem cells, we investigated whether hypoxia similarly contributes to LSC persistence. Importantly, we found that although breakpoint cluster region (BCR)-ABL1 kinase remained effectively inhibited by imatinib under hypoxia, apoptosis became partially suppressed. Furthermore, hypoxia enhanced the clonogenicity of CML cells, as well as their efficiency in repopulating immunodeficient mice, both in the presence and absence of imatinib. Hypoxia-inducible factor 1 ? (HIF1-?), which is the master regulatorof thehypoxia transcriptional response, is expressed in the BM specimens of CML individuals. Invitro, HIF1-? is stabilized during hypoxia, and its expression and transcriptional activity can be partially attenuated by concurrent imatinib treatment. Expression analysis demonstrates at the whole-transcriptome level that hypoxia and imatinib regulate distinct subsets of genes. Functionally, knockdown of HIF1-? abolished the enhanced clonogenicity during hypoxia. Taken together, our results suggest that in the hypoxic microenvironment, HIF1-? signaling supports LSC persistence independent of BCR-ABL1 kinase activity. Thus, targeting HIF1-? and its pathway components may be therapeutically important for the complete eradication of LSCs. � 2014 by The American Society of Hematology.
dc.publisherAmerican Society of Hematology
dc.sourceScopus
dc.typeArticle
dc.contributor.departmentMEDICINE
dc.contributor.departmentPATHOLOGY
dc.contributor.departmentDUKE-NUS MEDICAL SCHOOL
dc.description.doi10.1182/blood-2013-07-511907
dc.description.sourcetitleBlood
dc.description.volume123
dc.description.issue21
dc.description.page3316-3326
dc.published.statePublished
Appears in Collections:Elements
Staff Publications

Show simple item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
Physiologic hypoxia promotes maintenance.pdf2.17 MBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.