Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 23(8): e54483, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35758159

ABSTRACT

DNA lesions occur across the genome and constitute a threat to cell viability; however, damage at specific genomic loci has a relatively greater impact on overall genome stability. The ribosomal RNA gene repeats (rDNA) are emerging fragile sites. Recent progress in understanding how the rDNA damage response is organized has highlighted a key role of adaptor proteins. Here, we show that the scaffold tumor suppressor RASSF1A is recruited to rDNA breaks. RASSF1A recruitment to double-strand breaks is mediated by 53BP1 and depends on RASSF1A phosphorylation at Serine 131 by ATM kinase. Employing targeted rDNA damage, we uncover that RASSF1A recruitment promotes local ATM signaling. RASSF1A silencing, a common epigenetic event during malignant transformation, results in persistent breaks, rDNA copy number alterations and decreased cell viability. Overall, we identify a novel role for RASSF1A at rDNA break sites, provide mechanistic insight into how the DNA damage response is organized in a chromatin context, and provide further evidence for how silencing of the RASSF1A tumor suppressor contributes to genome instability.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Binding Proteins , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , DNA Repair , DNA, Ribosomal/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Phosphorylation , Signal Transduction/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
2.
Cancer Cell ; 37(5): 690-704.e8, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32330454

ABSTRACT

Acute erythroid leukemia (AEL) commonly involves both myeloid and erythroid lineage transformation. However, the mutations that cause AEL and the cell(s) that sustain the bilineage leukemia phenotype remain unknown. We here show that combined biallelic Cebpa and Gata2 zinc finger-1 (ZnF1) mutations cooperatively induce bilineage AEL, and that the major leukemia-initiating cell (LIC) population has a neutrophil-monocyte progenitor (NMP) phenotype. In pre-leukemic NMPs Cebpa and Gata2 mutations synergize by increasing erythroid transcription factor (TF) expression and erythroid TF chromatin access, respectively, thereby installing ectopic erythroid potential. This erythroid-permissive chromatin conformation is retained in bilineage LICs. These results demonstrate that synergistic transcriptional and epigenetic reprogramming by leukemia-initiating mutations can generate neomorphic pre-leukemic progenitors, defining the lineage identity of the resulting leukemia.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Lineage , Cell Transformation, Neoplastic/pathology , Erythroid Precursor Cells/pathology , GATA2 Transcription Factor/genetics , Leukemia, Erythroblastic, Acute/pathology , Mutation , Neutrophils/pathology , Aged , Alleles , Animals , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Erythroid Precursor Cells/metabolism , Female , GATA1 Transcription Factor/genetics , Humans , Leukemia, Erythroblastic, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophils/metabolism , Zinc Fingers
3.
Nat Immunol ; 19(1): 85-97, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29167569

ABSTRACT

The hierarchy of human hemopoietic progenitor cells that produce lymphoid and granulocytic-monocytic (myeloid) lineages is unclear. Multiple progenitor populations produce lymphoid and myeloid cells, but they remain incompletely characterized. Here we demonstrated that lympho-myeloid progenitor populations in cord blood - lymphoid-primed multi-potential progenitors (LMPPs), granulocyte-macrophage progenitors (GMPs) and multi-lymphoid progenitors (MLPs) - were functionally and transcriptionally distinct and heterogeneous at the clonal level, with progenitors of many different functional potentials present. Although most progenitors had the potential to develop into only one mature cell type ('uni-lineage potential'), bi- and rarer multi-lineage progenitors were present among LMPPs, GMPs and MLPs. Those findings, coupled with single-cell expression analyses, suggest that a continuum of progenitors execute lymphoid and myeloid differentiation, rather than only uni-lineage progenitors' being present downstream of stem cells.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling/methods , Lymphoid Progenitor Cells/metabolism , Myeloid Progenitor Cells/metabolism , Single-Cell Analysis/methods , Animals , Cell Lineage/genetics , Cell Separation/methods , Cells, Cultured , Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation/methods , Humans , Mice , Transplantation, Heterologous
4.
Stem Cells ; 35(2): 299-310, 2017 02.
Article in English | MEDLINE | ID: mdl-27859962

ABSTRACT

Molecular mechanisms governing maintenance, commitment, and differentiation of stem cells are largely unexploited. Molecules involved in the regulation of multiple cellular processes are of particular importance for stem cell physiology, as they integrate different signals and coordinate cellular decisions related with self-renewal and fate determination. Geminin has emerged as a critical factor in DNA replication and stem cell differentiation in different stem cell populations. Its inhibitory interaction with Cdt1, a member of the prereplicative complex, ensures the controlled timing of DNA replication and, consequently, genomic stability in actively proliferating cells. In embryonic as well as somatic stem cells, Geminin has been shown to interact with transcription factors and epigenetic regulators to drive gene expression programs and ultimately guide cell fate decisions. An ever-growing number of studies suggests that these interactions of Geminin and proteins regulating transcription are conserved among metazoans. Interactions between Geminin and proteins modifying the epigenome, such as members of the repressive Polycomb group and the SWI/SNF proteins of the permissive Trithorax, have long been established. The complexity of these interactions, however, is only just beginning to unravel, revealing key roles on maintaining stem cell self-renewal and fate specification. In this review, we summarize current knowledge and give new perspectives for the role of Geminin on transcriptional and epigenetic regulation, alongside with its regulatory activity in DNA replication and their implication in the regulation of stem and progenitor cell biology. Stem Cells 2017;35:299-310.


Subject(s)
DNA Replication/genetics , Epigenesis, Genetic , Geminin/metabolism , Stem Cells/metabolism , Transcription, Genetic , Animals , Genomic Instability , Humans
5.
J Exp Med ; 213(8): 1513-35, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27377587

ABSTRACT

Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34(-), there are multiple, nonhierarchically arranged CD34(+) and CD34(-) LSC populations. Within CD34(-) and CD34(+) LSC-containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34(-) LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34(-) mature granulocyte-macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.


Subject(s)
Antigens, CD34/genetics , Granulocyte-Macrophage Progenitor Cells/metabolism , Leukemia, Myeloid, Acute , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/pathology
6.
Data Brief ; 7: 889-93, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27077091

ABSTRACT

We performed cDNA microarrays (Affymetrix Mouse Gene 1.0 ST Chip) to analyze the transcriptome of hematopoietic stem and progenitor cells (HSPCs) from E15.5dpc wild type and Geminin (Gmnn) knockout embryos. Lineage negative cells from embryonic livers were isolated using fluorescence activated cell sorting. RNA samples were used to examine the transcriptional programs regulated by Geminin during embryonic hematopoiesis. The data sets were analyzed using the GeneSpring v12.5 platform (Agilent). The list of differentially expressed genes was filtered in meta-analyses to investigate the molecular basis of the phenotype observed in the knockout embryos, which exhibited defective hematopoiesis and death. The data from this study are related to the research article "Geminin deletion increases the number of fetal hematopoietic stem cells by affecting the expression of key transcription factors" (Karamitros et al., 2015) [1]. The microarray dataset has been deposited at the Gene Expression Omnibus (GEO) under accession GEO: GSE53056.

7.
Blood Adv ; 1(3): 160-169, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-29296933

ABSTRACT

Chronic myeloid leukemia (CML) is an excellent model of the multistep processes in cancer. Initiating BCR-ABL mutations are required for the initial phase of the disease (chronic phase, CP-CML). Some CP-CML patients acquire additional mutation(s) that transforms CP-CML to poor prognosis, hard to treat, acute myeloid or lymphoid leukemia or blast phase CML (BP-CML). It is unclear where in the hemopoietic hierarchy additional mutations are acquired in BP-CML, how the hemopoietic hierarchy is altered as a consequence, and the cellular identity of the resulting leukemia-propagating stem cell (LSC) populations. Here, we show that myeloid BP-CML is associated with expanded populations that have the immunophenotype of normal progenitor populations that vary between patients. Serial transplantation in immunodeficient mice demonstrated functional LSCs reside in multiple populations with the immunophenotype of normal progenitor as well as stem cells. Multicolor fluorescence in situ hybridization detected serial acquisition of cytogenetic abnormalities of chromosome 17, associated with transformation to BP-CML, that is detected with equal frequency in all functional LSC compartments. New effective myeloid BP-CML therapies will likely have to target all these LSC populations.

8.
Development ; 142(1): 70-81, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25516969

ABSTRACT

Balancing stem cell self-renewal and initiation of lineage specification programs is essential for the development and homeostasis of the hematopoietic system. We have specifically ablated geminin in the developing murine hematopoietic system and observed profound defects in the generation of mature blood cells, leading to embryonic lethality. Hematopoietic stem cells (HSCs) accumulated in the fetal liver following geminin ablation, while committed progenitors were reduced. Genome-wide transcriptome analysis identified key HSC transcription factors as being upregulated upon geminin deletion, revealing a gene network linked with geminin that controls fetal hematopoiesis. In order to obtain mechanistic insight into the ability of geminin to regulate transcription, we examined Hoxa9 as an example of a key gene in definitive hematopoiesis. We demonstrate that in human K562 cells geminin is associated with HOXA9 regulatory elements and its absence increases HOXA9 transcription similarly to that observed in vivo. Moreover, silencing geminin reduced recruitment of the PRC2 component SUZ12 to the HOXA9 locus and resulted in an increase in RNA polymerase II recruitment and H3K4 trimethylation (H3K4me3), whereas the repressive marks H3K9me3 and H3K27me3 were reduced. The chromatin landscape was also modified at the regulatory regions of HOXA10 and GATA1. K562 cells showed a reduced ability to differentiate to erythrocytes and megakaryocytes upon geminin silencing. Our data suggest that geminin is indispensable for fetal hematopoiesis and regulates the generation of a physiological pool of stem and progenitor cells in the fetal hematopoietic system.


Subject(s)
Fetus/cytology , Geminin/deficiency , Gene Deletion , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Transcription Factors/genetics , Animals , Cell Count , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryo Loss/metabolism , Embryo Loss/pathology , Epigenesis, Genetic , Geminin/metabolism , Gene Ontology , Genetic Loci , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Liver/cytology , Liver/embryology , Mice , Neoplasm Proteins , Polycomb Repressive Complex 2/metabolism , Protein Processing, Post-Translational , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Transcriptome/genetics
9.
Mech Ageing Dev ; 134(1-2): 10-23, 2013.
Article in English | MEDLINE | ID: mdl-23142824

ABSTRACT

Cellular senescence is a permanent out-of-cycle state regulated by molecular circuits acting during the G1 phase of the cell cycle. Cdt1 is a central regulator of DNA replication licensing acting during the G1 phase and it is negatively controlled by Geminin. Here, we characterize the cell cycle expression pattern of Cdt1 and Geminin during successive passages of primary fibroblasts and compare it to tumour-derived cell lines. Cdt1 and Geminin are strictly expressed in distinct subpopulations of young fibroblasts, similarly to cancer cells, with Geminin accumulating shortly after the onset of S phase. Cdt1 and Geminin are down-regulated when primary human and mouse fibroblasts undergo replicative or stress-induced senescence. RNAi-mediated Geminin knock-down in human cells enhances the appearance of phenotypic and molecular features of senescence. Mouse embryonic fibroblasts heterozygous for Geminin exhibit accelerated senescence compared to control fibroblasts. In contrast, ectopic expression of Geminin in mouse embryonic fibroblasts delays the appearance of the senescent phenotype. Taken together, our data suggest that changes in Geminin expression levels affect the establishment of senescence pathways.


Subject(s)
Cellular Senescence/physiology , Fibroblasts/metabolism , G1 Phase/physiology , Gene Expression Regulation/physiology , S Phase/physiology , SMN Complex Proteins/biosynthesis , Animals , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , HeLa Cells , Humans , Mice , SMN Complex Proteins/genetics
10.
Crit Rev Immunol ; 31(3): 209-31, 2011.
Article in English | MEDLINE | ID: mdl-21740351

ABSTRACT

Thymic T cell differentiation to peripheral T cells aims to assist the generation of effector cells mediating adaptive immune responses. During this process, which takes place during embryogenesis and in adulthood, proliferation is coupled with changes in chromatin organization and transcription. Moreover, B and T lymphocytes start to proliferate and rapidly expand their numbers when activated following an encounter with an antigen. This expansion phase is accompanied by differentiation of naïve T cells and is followed by a period of population contraction, resulting in only a small fraction of the expanded population surviving and entering the memory cell pool. The kinetics of the expansion and contraction affect the speed of antigen clearance and the clinical course of disease. Molecules that are involved in the coordination of proliferation, chromatin reorganization, and transcriptional regulation are likely to play an important role in T cell generation, homeostasis, and disease. Here we review how cell cycle regulators affect lymphoid system development and homeostasis and discuss recent evidence implicating the cell cycle inhibitor Geminin in this process. Geminin has been shown to coordinate proliferation and differentiation by regulating cell cycle progression, chromatin organization, and transcription in the nervous system. In the immune system, progenitor T cell commitment and differentiation progresses normally in the absence of Geminin. However, Geminin is required for TCR response in vitro and T cell proliferation upon lymphopenia-induced proliferation, suggesting that Geminin might be an essential factor for T cell expansion during the immune response.


Subject(s)
Cell Cycle Proteins , Cell Cycle/immunology , Cell Differentiation , Chromatin Assembly and Disassembly/immunology , Homeostasis/immunology , Immunity , T-Lymphocytes/immunology , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Proliferation , Chromatin/genetics , Chromatin/immunology , Chromatin Assembly and Disassembly/genetics , Epigenesis, Genetic/immunology , Geminin , Homeostasis/genetics , Humans , Immunity/genetics , Immunologic Memory , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcriptional Activation
11.
Cell Cycle ; 9(16): 3181-5, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20697201

ABSTRACT

The interplay of proliferation and differentiation is essential for normal development and organogenesis. Geminin is a cell cycle regulator which controls licensing of origins for DNA replication, safeguarding genomic stability. Geminin has also been shown to regulate cellular decisions of self-renewal versus commitment of neuronal progenitor cells. We discuss here our recent analysis of mice with conditional inactivation of the Geminin gene in the immune system. Our data indicate that Geminin is not indispensable for every cell division: in the absence of Geminin, development of progenitor T cells appears largely unaffected. In contrast, rapid cell divisions, taking place in vitro upon TCR receptor activation or in vivo during homeostatic proliferation, are defective.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cell Differentiation , Cell Proliferation , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , G1 Phase , Geminin , Genomic Instability , Mice , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Polycomb-Group Proteins , Repressor Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factors/metabolism
12.
J Immunol ; 184(5): 2432-41, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20107189

ABSTRACT

Stem/progenitor cells coordinate proliferation and differentiation, giving rise to appropriate cell numbers of functionally specialized cells during organogenesis. In different experimental systems, Geminin was shown to maintain progenitor cells and participate in fate determination decisions and organogenesis. Although the exact mechanisms are unclear, Geminin has been postulated to influence proliferation versus differentiation decisions. To gain insight into the in vivo role of Geminin in progenitor cell division and differentiation, we have generated mice that specifically lack Geminin in cells of lymphoid lineage through Cre-mediated recombination. T cells lacking Geminin expression upregulate early activation markers efficiently upon TCR stimulation in vitro and are able to enter the S phase of cell cycle, but show a marked defect in completing the cycle, leading to a large proportion of T cells accumulating in S/G2/M phases. Accordingly, T cells deficient in Geminin show a reduced ability to repopulate lymphopenic hosts in vivo. Contrary to expectations, Geminin deficiency does not alter development and differentiation of T cells in vivo. Our data suggest that Geminin is required for the proliferation events taking place either in vitro upon TCR receptor activation or during homeostatic expansion, but appears to be redundant for the proliferation and differentiation of the majority of progenitor T cell populations.


Subject(s)
Cell Cycle Proteins/physiology , Cell Proliferation , Nuclear Proteins/physiology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/genetics , Cell Division/physiology , Cell Lineage , Cells, Cultured , DNA-Binding Proteins/metabolism , Flow Cytometry , G2 Phase/genetics , G2 Phase/physiology , Geminin , Homeostasis/genetics , Homeostasis/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , S Phase/genetics , S Phase/physiology , Spleen/cytology , Spleen/metabolism , T-Lymphocytes/metabolism , Thymus Gland/metabolism
13.
Front Biosci ; 13: 4485-94, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508524

ABSTRACT

Cdt1 and its inhibitor Geminin are important regulators of replication licensing. In normal cells, a critical balance between these two proteins ensures that firing of each origin along the genome will take place only once per cell cycle. Cdt1 overexpression in cell lines and animals leads to aberrant replication, activates DNA damage checkpoints and predisposes for malignant transformation. Geminin inactivation mimics the effects of Cdt1 overexpression in cells and generates mitotic defects and abnormal chromosome segregation. Aberrant expression of Cdt1 and Geminin is thus linked to DNA replication defects, aneuploidy and genomic instability. These traits are considered integral to precancerous states and essential elements for malignant transformation. Moreover, Cdt1 and Geminin expression is deregulated in human tumor specimens and Cdt1 and Geminin may represent novel markers useful for cancer diagnosis and prognosis.


Subject(s)
Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , DNA Replication , Neoplasms/genetics , Antigens, CD/analysis , CD11 Antigens/analysis , Geminin , Gene Expression Regulation, Neoplastic , Genetic Markers , Humans , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...