Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 7: 12475, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27534895

ABSTRACT

In acute myeloid leukaemia (AML) initiating pre-leukaemic lesions can be identified through three major hallmarks: their early occurrence in the clone, their persistence at relapse and their ability to initiate multilineage haematopoietic repopulation and leukaemia in vivo. Here we analyse the clonal composition of a series of AML through these characteristics. We find that not only DNMT3A mutations, but also TET2, ASXL1 mutations, core-binding factor and MLL translocations, as well as del(20q) mostly fulfil these criteria. When not eradicated by AML treatments, pre-leukaemic cells with these lesions can re-initiate the leukaemic process at various stages until relapse, with a time-dependent increase in clonal variegation. Based on the nature, order and association of lesions, we delineate recurrent genetic hierarchies of AML. Our data indicate that first lesions, variegation and treatment selection pressure govern the expansion and adaptive behaviour of the malignant clone, shaping AML in a time-dependent manner.


Subject(s)
Clonal Evolution/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Animals , Base Sequence , Clone Cells , Epigenesis, Genetic , Gene Rearrangement/genetics , Hematopoiesis , Humans , Mice , Mutation/genetics , Single-Cell Analysis , Time Factors
2.
Stem Cell Reports ; 3(6): 1085-102, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25458892

ABSTRACT

Hematopoietic stem cells (HSCs) are characterized by the capacity for self-renewal and the ability to reconstitute the entire hematopoietic compartment. Thrombopoietin maintains adult HSCs in a quiescent state through the induction of cell cycle inhibitors p57(Kip2) and p19(INK4d). Using the p19(INK4d-/-) mouse model, we investigated the role of p19(INK4d) in basal and stress-induced hematopoiesis. We demonstrate that p19(INK4d) is involved in the regulation of HSC quiescence by inhibition of the G0/G1 cell cycle transition. Under genotoxic stress conditions, the absence of p19(INK4d) in HSCs leads to accelerated cell cycle exit, accumulation of DNA double-strand breaks, and apoptosis when cells progress to the S/G2-M stages of the cell cycle. Moreover, p19(INK4d) controls the HSC microenvironment through negative regulation of megakaryopoiesis. Deletion of p19(INK4d) results in megakaryocyte hyperproliferation and increased transforming growth factor ß1 secretion. This leads to fibrosis in the bone marrow and spleen, followed by loss of HSCs during aging.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p19/genetics , DNA Damage , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/genetics , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Count , Cell Differentiation/genetics , Cell Lineage/genetics , Cyclin-Dependent Kinase Inhibitor p19/deficiency , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Hematopoiesis , Mice , Mice, Knockout , Osteosclerosis/genetics , Osteosclerosis/pathology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Resting Phase, Cell Cycle/genetics , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL