Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 9(1): 1281, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29599503

ABSTRACT

Nitric oxide (NO) synthesis is a late event during differentiation of mouse embryonic stem cells (mESC) and occurs after release from serum and leukemia inhibitory factor (LIF). Here we show that after release from pluripotency, a subpopulation of mESC, kept in the naive state by 2i/LIF, expresses endothelial nitric oxide synthase (eNOS) and endogenously synthesizes NO. This eNOS/NO-positive subpopulation (ESNO+) expresses mesendodermal markers and is more efficient in the generation of cardiovascular precursors than eNOS/NO-negative cells. Mechanistically, production of endogenous NO triggers rapid Hdac2 S-nitrosylation, which reduces association of Hdac2 with the transcriptional repression factor Zeb1, allowing mesendodermal gene expression. In conclusion, our results suggest that the interaction between Zeb1, Hdac2, and eNOS is required for early mesendodermal differentiation of naive mESC.


Subject(s)
Histone Deacetylase 2/metabolism , Mouse Embryonic Stem Cells/cytology , Myocardium/cytology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , HeLa Cells , Humans , Leukemia Inhibitory Factor/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myocardium/metabolism
2.
Stem Cells ; 34(3): 699-710, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26731607

ABSTRACT

Hematopoietic stem cells (HSCs) maintain blood cell production life-long by their unique abilities of self-renewal and differentiation into all blood cell lineages. Growth arrest and DNA-damage-inducible 45 alpha (GADD45A) is induced by genotoxic stress in HSCs. GADD45A has been implicated in cell cycle control, cell death and senescence, as well as in DNA-damage repair. In general, GADD45A provides cellular stability by either arresting the cell cycle progression until DNA damage is repaired or, in cases of fatal damage, by inducing apoptosis. However, the function of GADD45A in hematopoiesis remains controversial. We revealed the changes in murine HSC fate control orchestrated by the expression of GADD45A at single cell resolution. In contrast to other cellular systems, GADD45A expression did not cause a cell cycle arrest or an alteration in the decision between cell survival and apoptosis in HSCs. Strikingly, GADD45A strongly induced and accelerated the differentiation program in HSCs. Continuous tracking of individual HSCs and their progeny via time-lapse microscopy elucidated that once GADD45A was expressed, HSCs differentiate into committed progenitors within 29 hours. GADD45A-expressing HSCs failed to long-term reconstitute the blood of recipients by inducing multilineage differentiation in vivo. Importantly, γ-irradiation of HSCs induced their differentiation by upregulating endogenous GADD45A. The differentiation induction by GADD45A was transmitted by activating p38 Mitogen-activated protein kinase (MAPK) signaling and allowed the generation of megakaryocytic-erythroid, myeloid, and lymphoid lineages. These data indicate that genotoxic stress-induced GADD45A expression in HSCs prevents their fatal transformation by directing them into differentiation and thereby clearing them from the system.


Subject(s)
Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Hematopoietic Stem Cells , Nuclear Proteins/genetics , Animals , Apoptosis/genetics , Cell Cycle Proteins/biosynthesis , Cell Proliferation/genetics , Cell Survival/genetics , DNA Damage/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Humans , Mice , Nuclear Proteins/biosynthesis , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics
3.
Nat Commun ; 6: 8928, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26603207

ABSTRACT

Haematopoietic stem cells (HSCs) require the right composition of microRNAs (miR) for proper life-long balanced blood regeneration. Here we show a regulatory circuit that prevents excessive HSC self-renewal by upregulation of miR-193b upon self-renewal promoting thrombopoietin (TPO)-MPL-STAT5 signalling. In turn, miR-193b restricts cytokine signalling, by targeting the receptor tyrosine kinase c-KIT. We generated a miR-193b knockout mouse model to unravel the physiological function of miR-193b in haematopoiesis. MiR-193b(-/-) mice show a selective gradual enrichment of functional HSCs, which are fully competent in multilineage blood reconstitution upon transplantation. The absence of miR-193b causes an accelerated expansion of HSCs, without altering cell cycle or survival, but by decelerating differentiation. Conversely, ectopic miR-193b expression restricts long-term repopulating HSC expansion and blood reconstitution. MiR-193b-deficient haematopoietic stem and progenitor cells exhibit increased basal and cytokine-induced STAT5 and AKT signalling. This STAT5-induced microRNA provides a negative feedback for excessive signalling to restrict uncontrolled HSC expansion.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Thrombopoietin/metabolism , STAT5 Transcription Factor/metabolism , Thrombopoietin/metabolism , Animals , Cell Proliferation/genetics , Flow Cytometry , Mice , Mice, Knockout , MicroRNAs/metabolism , Receptors, Cytokine/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time-Lapse Imaging
4.
Stem Cell Reports ; 3(1): 34-43, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25068120

ABSTRACT

The balance of self-renewal and differentiation in long-term repopulating hematopoietic stem cells (LT-HSC) must be strictly controlled to maintain blood homeostasis and to prevent leukemogenesis. Hematopoietic cytokines can induce differentiation in LT-HSCs; however, the molecular mechanism orchestrating this delicate balance requires further elucidation. We identified the tumor suppressor GADD45G as an instructor of LT-HSC differentiation under the control of differentiation-promoting cytokine receptor signaling. GADD45G immediately induces and accelerates differentiation in LT-HSCs and overrides the self-renewal program by specifically activating MAP3K4-mediated MAPK p38. Conversely, the absence of GADD45G enhances the self-renewal potential of LT-HSCs. Videomicroscopy-based tracking of single LT-HSCs revealed that, once GADD45G is expressed, the development of LT-HSCs into lineage-committed progeny occurred within 36 hr and uncovered a selective lineage choice with a severe reduction in megakaryocytic-erythroid cells. Here, we report an unrecognized role of GADD45G as a central molecular linker of extrinsic cytokine differentiation and lineage choice control in hematopoiesis.


Subject(s)
Cytokines/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cell Differentiation/physiology , Flow Cytometry , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinase 4/genetics , MAP Kinase Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Video , GADD45 Proteins
5.
Blood ; 105(5): 2107-14, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15536149

ABSTRACT

Activating fetal liver tyrosine kinase 3 (Flt3) mutations represent the most common genetic aberrations in acute myeloid leukemia (AML). Most commonly, they occur as internal tandem duplications in the juxtamembrane domain (Flt3-ITD) that transform myeloid cells in vitro and in vivo and that induce aberrant signaling and biologic functions. We identified RGS2, a regulator of G-protein signaling, as a gene specifically repressed by Flt3-ITD. Here we demonstrate an important role of RGS2 in Flt3-ITD-mediated transformation. RGS2 was repressed after forced expression of activating Flt3 mutations in 2 myeloid cell lines (32Dcl3 and NB4). Furthermore, RGS2 was repressed in Flt3-mutation-positive AML cases in comparison to Flt3-mutation-negative cases, especially in Flt3-ITD-positive cases with a high ITD-to-wild-type (WT) ratio. Coexpression of RGS2 with Flt3-ITD inhibited Flt3-ITD-induced autonomous proliferation and clonal growth of 32D cells. RGS2 also inhibited Flt3-ITD-induced phosphorylation of Akt and glycogen synthase kinase beta (Gsk3-beta) without influencing signal transducer and activator of transcription 5 (STAT5) activation. In addition, RGS2 reinduced the expression of Flt3-ITD-repressed CCAAT/enhancer-binding protein alpha (c/EBPalpha) and antagonized the Flt3-ITD-induced differentiation block in 32D cells. Expression analyses in myeloid cell lines revealed induction of RGS2 during granulocytic but not during monocytic differentiation. Taken together, RGS2 is a novel mediator of myeloid differentiation, and its repression is an important event in Flt3-ITD-induced transformation.


Subject(s)
Cell Transformation, Neoplastic , Leukemia, Myeloid/genetics , Mutation , Myeloid Cells/pathology , Proto-Oncogene Proteins/genetics , RGS Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Acute Disease , Cell Differentiation , Cell Line , Cell Proliferation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HL-60 Cells , Humans , Leukemia, Myeloid/etiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Repressor Proteins , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...