Egyszerű nézet

dc.contributor.author Wang LD
dc.contributor.author Ficarro SB
dc.contributor.author Hutchinson JN
dc.contributor.author Csépányi-Kömi Roland
dc.contributor.author Nguyen PT
dc.contributor.author Wisniewski Éva
dc.contributor.author Sullivan J
dc.contributor.author Hofmann O
dc.contributor.author Ligeti Erzsébet
dc.contributor.author Marto JA
dc.contributor.author Wagers AJ
dc.date.accessioned 2018-10-02T09:52:36Z
dc.date.available 2018-10-02T09:52:36Z
dc.date.issued 2016
dc.identifier 84993960865
dc.identifier.citation pagination=1465-1474; journalVolume=128; journalIssueNumber=11; journalTitle=BLOOD;
dc.identifier.uri http://repo.lib.semmelweis.hu//handle/123456789/4814
dc.identifier.uri doi:10.1182/blood-2016-05-711424
dc.description.abstract Protein phosphorylation is a central mechanism of signal transduction that both positively and negatively regulates protein function. Large-scale studies of the dynamic phosphorylation states of cell signaling systems have been applied extensively in cell lines and whole tissues to reveal critical regulatory networks, and candidate-based evaluations of phosphorylation in rare cell populations have also been informative. However, application of comprehensive profiling technologies to adult stem cell and progenitor populations has been challenging, due in large part to the scarcity of such cells in adult tissues. Here, we combine multi-color flow cytometry with highly efficient three dimensional HPLC/mass spectrometry to enable quantitative phosphoproteomic analysis from 200,000 highly purified primary mouse hematopoietic stem and progenitor cells (HSPCs). Using this platform, we identify ARHGAP25 as a novel regulator of HSPC mobilization and demonstrate that ARHGAP25 phosphorylation at serine 363 is an important modulator of its function. Our approach provides a robust platform for large-scale phosphoproteomic analyses performed with limited numbers of rare progenitor cells. Data from our study comprises a new resource for understanding the molecular signaling networks that underlie hematopoietic stem cell mobilization.
dc.relation.ispartof urn:issn:0006-4971
dc.title Phosphoproteomic profiling of mouse primary HSPCs reveals new regulators of HSPC mobilization
dc.type Journal Article
dc.date.updated 2018-02-16T12:47:28Z
dc.language.rfc3066 en
dc.identifier.mtmt 3105028
dc.identifier.wos 000385733200005
dc.identifier.pubmed 27365422
dc.mtmt.swordnote FELTÖLTŐ: Sonnevend Kinga - sonnevend.kinga@med.semmelweis-univ.hu


Kapcsolódó fájlok:

A fájl jelenleg csak egyetemi IP címről érhető el.

Megtekintés/Megnyitás

Ez a rekord az alábbi gyűjteményekben szerepel:

Egyszerű nézet