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

Database
Language
Affiliation country
Publication year range
1.
Haematologica ; 107(3): 680-689, 2022 03 01.
Article in English | MEDLINE | ID: mdl-33691379

ABSTRACT

Recurrent loss-of-function mutations of spliceosome gene, ZRSR2, occur in myelodysplastic syndromes (MDS). Mutation/loss of ZRSR2 in human myeloid cells primarily causes impaired splicing of the U12-type introns. In order to further investigate the role of this splice factor in RNA splicing and hematopoietic development, we generated mice lacking ZRSR2. Unexpectedly, Zrsr2-deficient mice developed normal hematopoiesis with no abnormalities in myeloid differentiation evident in either young or ≥1-year old knockout mice. Repopulation ability of Zrsr2-deficient hematopoietic stem cells was also unaffected in both competitive and non-competitive reconstitution assays. Myeloid progenitors lacking ZRSR2 exhibited mis-splicing of U12-type introns, however, this phenotype was moderate compared to the ZRSR2-deficient human cells. Our investigations revealed that a closely related homolog, Zrsr1, expressed in the murine hematopoietic cells, but not in human cells contributes to splicing of U12-type introns. Depletion of Zrsr1 in Zrsr2 KO myeloid cells exacerbated retention of the U12-type introns, thus highlighting a collective role of ZRSR1 and ZRSR2 in murine U12-spliceosome. We also demonstrate that aberrant retention of U12-type introns of MAPK9 and MAPK14 leads to their reduced protein expression. Overall, our findings highlight that both ZRSR1 and ZRSR2 are functional components of the murine U12-spliceosome, and depletion of both proteins is required to accurately model ZRSR2-mutant MDS in mice.


Subject(s)
Myelodysplastic Syndromes , RNA Splicing , Ribonucleoproteins , Splicing Factor U2AF , Animals , Introns , Mice , Mutation , Myelodysplastic Syndromes/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Spliceosomes/genetics , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
2.
Haematologica ; 103(8): 1269-1277, 2018 08.
Article in English | MEDLINE | ID: mdl-29773596

ABSTRACT

Maturation of granulocytes is dependent on controlled gene expression by myeloid lineage restricted transcription factors. CEBPE is one of the essential transcription factors required for granulocytic differentiation. Identification of downstream targets of CEBPE is vital to understand better its role in terminal granulopoiesis. In this study, we have identified Card10 as a novel target of CEBPE. We show that CEBPE binds to regulatory elements upstream of the murine Card10 locus, and expression of CARD10 is significantly reduced in Cebpe knock-out mice. Silencing Card10 in a human cell line and in murine primary cells impaired granulopoiesis, affecting expression of genes involved in myeloid cell development and function. Taken together, our data demonstrate for the first time that Card10 is expressed in granulocytes and is a direct target of CEBPE with functions extending to myeloid differentiation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation , Granulocytes/cytology , Animals , Binding Sites , Cell Line , Cells, Cultured , Gene Expression Regulation , Granulocytes/metabolism , Humans , Mice , Myeloid Cells , Protein Binding , Transcription Factors/genetics
3.
Haematologica ; 103(12): 1980-1990, 2018 12.
Article in English | MEDLINE | ID: mdl-30093396

ABSTRACT

Chromosomal translocation t(8;21)(q22;q22) which leads to the generation of oncogenic RUNX1-RUNX1T1 (AML1-ETO) fusion is observed in approximately 10% of acute myelogenous leukemia (AML). To identify somatic mutations that co-operate with t(8;21)-driven leukemia, we performed whole and targeted exome sequencing of an Asian cohort at diagnosis and relapse. We identified high frequency of truncating alterations in ASXL2 along with recurrent mutations of KIT, TET2, MGA, FLT3, and DHX15 in this subtype of AML. To investigate in depth the role of ASXL2 in normal hematopoiesis, we utilized a mouse model of ASXL2 deficiency. Loss of ASXL2 caused progressive hematopoietic defects characterized by myeloid hyperplasia, splenomegaly, extramedullary hematopoiesis, and poor reconstitution ability in transplantation models. Parallel analyses of young and >1-year old Asxl2-deficient mice revealed age-dependent perturbations affecting, not only myeloid and erythroid differentiation, but also maturation of lymphoid cells. Overall, these findings establish a critical role for ASXL2 in maintaining steady state hematopoiesis, and provide insights into how its loss primes the expansion of myeloid cells.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Hematopoiesis/genetics , Myeloid Cells/metabolism , Repressor Proteins/genetics , Acute Disease , Animals , Gene Expression Profiling/methods , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis/genetics
4.
Blood Adv ; 7(21): 6553-6566, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37611161

ABSTRACT

The adenosine triphosphate (ATP)-dependent chromatin remodeling complex, SWItch/Sucrose Non-Fermentable (SWI/SNF), has been implicated in normal hematopoiesis. The AT-rich interaction domain 1B (ARID1B) and its paralog, ARID1A, are mutually exclusive, DNA-interacting subunits of the BRG1/BRM-associated factor (BAF) subclass of SWI/SNF complex. Although the role of several SWI/SNF components in hematopoietic differentiation and stem cell maintenance has been reported, the function of ARID1B in hematopoietic development has not been defined. To this end, we generated a mouse model of Arid1b deficiency specifically in the hematopoietic compartment. Unlike the extensive phenotype observed in mice deficient in its paralog, ARID1A, Arid1b knockout (KO) mice exhibited a modest effect on steady-state hematopoiesis. Nonetheless, transplantation experiments showed that the reconstitution of myeloid cells in irradiated recipient mice was dependent on ARID1B. Furthermore, to assess the effect of the complete loss of ARID1 proteins in the BAF complex, we generated mice lacking both ARID1A and ARID1B in the hematopoietic compartment. The double-KO mice succumbed to acute bone marrow failure resulting from complete loss of BAF-mediated chromatin remodeling activity. Our Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) analyses revealed that >80% of loci regulated by ARID1B were distinct from those regulated by ARID1A; and ARID1B controlled expression of genes crucial in myelopoiesis. Overall, loss of ARID1B affected chromatin dynamics in murine hematopoietic stem and progenitor cells, albeit to a lesser extent than cells lacking ARID1A.


Subject(s)
Hematopoiesis , Nuclear Proteins , Animals , Mice , Cell Differentiation/genetics , Chromatin , Hematopoiesis/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
5.
Leukemia ; 33(9): 2291-2305, 2019 09.
Article in English | MEDLINE | ID: mdl-30858552

ABSTRACT

Precise regulation of chromatin architecture is vital to physiological processes including hematopoiesis. ARID1A is a core component of the mammalian SWI/SNF complex, which is one of the ATP-dependent chromatin remodeling complexes. To uncover the role of ARID1A in hematopoietic development, we utilized hematopoietic cell-specific deletion of Arid1a in mice. We demonstrate that ARID1A is essential for maintaining the frequency and function of hematopoietic stem cells and its loss impairs the differentiation of both myeloid and lymphoid lineages. ARID1A deficiency led to a global reduction in open chromatin and ensuing transcriptional changes affected key genes involved in hematopoietic development. We also observed that silencing of ARID1A affected ATRA-induced differentiation of NB4 cells, suggesting its role in granulocytic differentiation of human leukemic cells. Overall, our study provides a comprehensive elucidation of the function of ARID1A in hematopoiesis and highlights the central role of ARID1A-containing SWI/SNF complex in maintaining chromatin dynamics in hematopoietic cells.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Cell Lineage , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL