Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Leukemia ; 33(6): 1411-1426, 2019 06.
Article in English | MEDLINE | ID: mdl-30679800

ABSTRACT

LSD1 has emerged as a promising epigenetic target in the treatment of acute myeloid leukemia (AML). We used two murine AML models based on retroviral overexpression of Hoxa9/Meis1 (H9M) or MN1 to study LSD1 loss of function in AML. The conditional knockout of Lsd1 resulted in differentiation with both granulocytic and monocytic features and increased ATRA sensitivity and extended the survival of mice with H9M-driven AML. The conditional knockout led to an increased expression of multiple genes regulated by the important myeloid transcription factors GFI1 and PU.1. These include the transcription factors GFI1B and IRF8. We also compared the effect of different irreversible and reversible inhibitors of LSD1 in AML and could show that only tranylcypromine derivatives were capable of inducing a differentiation response. We employed a conditional knock-in model of inactive, mutant LSD1 to study the effect of only interfering with LSD1 enzymatic activity. While this was sufficient to initiate differentiation, it did not result in a survival benefit in mice. Hence, we believe that targeting both enzymatic and scaffolding functions of LSD1 is required to efficiently treat AML. This finding as well as the identified biomarkers may be relevant for the treatment of AML patients with LSD1 inhibitors.


Subject(s)
Cell Differentiation/drug effects , DNA-Binding Proteins/metabolism , Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Tranylcypromine/pharmacology , Animals , Antidepressive Agents/pharmacology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Demethylases/physiology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Knockout , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Cells, Cultured
4.
Haematologica ; 103(4): 614-625, 2018 04.
Article in English | MEDLINE | ID: mdl-29326122

ABSTRACT

Differentiation of hematopoietic stem cells is regulated by a concert of different transcription factors. Disturbed transcription factor function can be the basis of (pre)malignancies such as myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Growth factor independence 1b (Gfi1b) is a repressing transcription factor regulating quiescence of hematopoietic stem cells and differentiation of erythrocytes and platelets. Here, we show that low expression of Gfi1b in blast cells is associated with an inferior prognosis of MDS and AML patients. Using different models of human MDS or AML, we demonstrate that AML development was accelerated with heterozygous loss of Gfi1b, and latency was further decreased when Gfi1b was conditionally deleted. Loss of Gfi1b significantly increased the number of leukemic stem cells with upregulation of genes involved in leukemia development. On a molecular level, we found that loss of Gfi1b led to epigenetic changes, increased levels of reactive oxygen species, as well as alteration in the p38/Akt/FoXO pathways. These results demonstrate that Gfi1b functions as an oncosuppressor in MDS and AML development.


Subject(s)
Leukemia, Myeloid, Acute/etiology , Myelodysplastic Syndromes/etiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Animals , Epigenomics , Forkhead Box Protein O1/metabolism , Gene Deletion , Heterozygote , Homozygote , Humans , Mice , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Sci Rep ; 7(1): 15720, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29147018

ABSTRACT

The differentiation of haematopoietic cells is regulated by a plethora of so-called transcription factors (TFs). Mutations in genes encoding TFs or graded reduction in their expression levels can induce the development of various malignant diseases such as acute myeloid leukaemia (AML). Growth Factor Independence 1 (GFI1) is a transcriptional repressor with key roles in haematopoiesis, including regulating self-renewal of haematopoietic stem cells (HSCs) as well as myeloid and lymphoid differentiation. Analysis of AML patients and different AML mouse models with reduced GFI1 gene expression levels revealed a direct link between low GFI1 protein level and accelerated AML development and inferior prognosis. Here, we report that upregulated expression of GFI1 in several widely used leukemic cell lines inhibits their growth and decreases the ability to generate colonies in vitro. Similarly, elevated expression of GFI1 impedes the in vitro expansion of murine pre-leukemic cells. Using a humanized AML model, we demonstrate that upregulation of GFI1 expression leads to myeloid differentiation morphologically and immunophenotypically, increased level of apoptosis and reduction in number of cKit+ cells. These results suggest that increasing GFI1 level in leukemic cells with low GFI1 expression level could be a therapeutic approach.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Transcription Factors/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Hematopoietic Stem Cells/metabolism , Humans , Mice, Inbred C57BL , Tumor Stem Cell Assay , Up-Regulation
6.
Haematologica ; 102(3): 484-497, 2017 03.
Article in English | MEDLINE | ID: mdl-28082345

ABSTRACT

Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.


Subject(s)
Cytoskeleton/metabolism , Integrins/metabolism , Megakaryocytes/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Actins/chemistry , Actins/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Profiling , Genetic Association Studies , Megakaryocytes/drug effects , Megakaryocytes/pathology , Megakaryocytes/ultrastructure , Mice , Mice, Knockout , Microtubules/metabolism , Phenotype , Platelet Count , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Transcriptome
7.
Exp Hematol ; 44(8): 713-726.e14, 2016 08.
Article in English | MEDLINE | ID: mdl-27216773

ABSTRACT

Epigenetic changes can contribute to development of acute myeloid leukemia (AML), a malignant disease of the bone marrow. A single-nucleotide polymorphism of transcription factor growth factor independence 1 (GFI1) generates a protein with an asparagine at position 36 (GFI1(36N)) instead of a serine at position 36 (GFI1(36S)), which is associated with de novo AML in humans. However, how GFI1(36N) predisposes to AML is poorly understood. To explore the mechanism, we used knock-in mouse strains expressing GFI1(36N) or GFI1(36S). Presence of GFI1(36N) shortened the latency and increased the incidence of AML in different murine models of myelodysplastic syndrome/AML. On a molecular level, GFI1(36N) induced genomewide epigenetic changes, leading to expression of AML-associated genes. On a therapeutic level, use of histone acetyltransferase inhibitors specifically impeded growth of GFI1(36N)-expressing human and murine AML cells in vitro and in vivo. These results establish, as a proof of principle, how epigenetic changes in GFI1(36N)-induced AML can be targeted.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Mutation , Transcription Factors/genetics , Amino Acid Substitution , Animals , Bone Marrow/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , Codon , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Models, Biological , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/mortality
8.
Blood ; 126(24): 2561-9, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26447191

ABSTRACT

The DNA-binding zinc finger transcription factors Gfi1 and Gfi1b were discovered more than 20 years ago and are recognized today as major regulators of both early hematopoiesis and hematopoietic stem cells. Both proteins function as transcriptional repressors by recruiting histone-modifying enzymes to promoters and enhancers of target genes. The establishment of Gfi1 and Gfi1b reporter mice made it possible to visualize their cell type-specific expression and to understand their function in hematopoietic lineages. We now know that Gfi1 is primarily important in myeloid and lymphoid differentiation, whereas Gfi1b is crucial for the generation of red blood cells and platelets. Several rare hematologic diseases are associated with acquired or inheritable mutations in the GFI1 and GFI1B genes. Certain patients with severe congenital neutropenia carry mutations in the GFI1 gene that lead to the disruption of the C-terminal zinc finger domains. Other mutations have been found in the GFI1B gene in families with inherited bleeding disorders. In addition, the Gfi1 locus is frequently found to be a proviral integration site in retrovirus-induced lymphomagenesis, and new, emerging data suggest a role of Gfi1 in human leukemia and lymphoma, underlining the role of both factors not only in normal hematopoiesis, but also in a wide spectrum of human blood diseases.


Subject(s)
DNA-Binding Proteins/physiology , Hematopoiesis/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Blood Cells/physiology , Congenital Bone Marrow Failure Syndromes , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Regulatory Networks , Genes, Reporter , Hematologic Diseases/genetics , Hematologic Neoplasms/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Histone Code/physiology , Humans , Leukemia Virus, Murine/physiology , Mice , Mice, Transgenic , Models, Molecular , Neutropenia/congenital , Neutropenia/genetics , Protein Conformation , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Virus Integration
9.
PLoS One ; 9(5): e96636, 2014.
Article in English | MEDLINE | ID: mdl-24800817

ABSTRACT

Growth factor independence 1b (GFI1B) is a DNA binding repressor of transcription with vital functions in hematopoiesis. Gfi1b-null embryos die at midgestation very likely due to defects in erythro- and megakaryopoiesis. To analyze the full functionality of Gfi1b, we used conditionally deficient mice that harbor floxed Gfi1b alleles and inducible (Mx-Cre, Cre-ERT) or erythroid specific (EpoR-Cre) Cre expressing transgenes. In contrast to the germline knockout, EpoR-Cre mediated erythroid specific ablation of Gfi1b allows full gestation, but causes perinatal lethality with very few mice surviving to adulthood. Both the embryonic deletion of Gfi1b by EpoR-Cre and the deletion in adult mice by Mx-Cre or Cre-ERT leads to reduced numbers of erythroid precursors, perturbed and delayed erythroid maturation, anemia and extramedullary erythropoiesis. Global expression analyses showed that the Hba-x, Hbb-bh1 and Hbb-y embryonic globin genes were upregulated in Gfi1b deficient TER119+ fetal liver cells over the gestation period from day 12.5-17.5 p.c. and an increased level of Hbb-bh1 and Hbb-y embryonic globin gene expression was even maintained in adult Gfi1b deficient mice. While the expression of Bcl11a, a regulator of embryonic globin expression was not affected by Gfi1b deficiency, the expression of Gata1 was reduced and the expression of Sox6, also involved in globin switch, was almost entirely lost when Gfi1b was absent. These findings establish Gfi1b as a regulator of embryonic globin expression and embryonic and adult erythroid maturation.


Subject(s)
Erythroid Cells/metabolism , Globins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Carrier Proteins/metabolism , DNA-Binding Proteins , Embryo, Mammalian/metabolism , Erythropoiesis , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Gene Knock-In Techniques , Globins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , SOXD Transcription Factors/metabolism
10.
Cancer Cell ; 23(2): 200-14, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23410974

ABSTRACT

Most patients with acute lymphoblastic leukemia (ALL) fail current treatments highlighting the need for better therapies. Because oncogenic signaling activates a p53-dependent DNA damage response and apoptosis, leukemic cells must devise appropriate countermeasures. We show here that growth factor independence 1 (Gfi1) can serve such a function because Gfi1 ablation exacerbates p53 responses and lowers the threshold for p53-induced cell death. Specifically, Gfi1 restricts p53 activity and expression of proapoptotic p53 targets such as Bax, Noxa (Pmaip1), and Puma (Bbc3). Subsequently, Gfi1 ablation cures mice from leukemia and limits the expansion of primary human T-ALL xenografts in mice. This suggests that targeting Gfi1 could improve the prognosis of patients with T-ALL or other lymphoid leukemias.


Subject(s)
Apoptosis , DNA Damage/genetics , DNA-Binding Proteins/physiology , Lymphoma, B-Cell/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , Animals , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays
11.
Am J Blood Res ; 2(4): 228-42, 2012.
Article in English | MEDLINE | ID: mdl-23226623

ABSTRACT

The transcriptional repressor Gfi1 regulates the expression of genes important for survival, proliferation and differentiation of hematopoietic cells. Gfi1 deficient mice are severely neutropenic and accumulate ill-defined CD11b(+)GR1(int) myeloid cells. Here we show that Gfi1 expression levels determine mono- or granulocytic lineage choice in precursor cells. In addition, we identify CD48 as a cell surface marker which enables a better definition of monocytes and granulocytes in mouse bone marrow. Using the CD48/Gr1/Gfi1 marker combination we can show that the CD11b(+)GR1(int) cells accumulating in Gfi1 deficient mice are monocytes and not granulocyte precursors. Expression of CD48, Gr1 and Gfi1 define different bone marrow subpopulations that are either committed to the granulocytic lineage, or bipotential precursors of granulocytes or monocytes. Finally, a comparison of genes differentially expressed between murine Gfi1 high granulocytic precursors and mature granulocytes with gene expression changes from human myeloblasts versus neutrophils show a strong resemblance of human and mouse differentiation pathways. This underlines the value of the markers CD48 and Gfi1 identified here to study human and murine granulo-monocytic differentiation.

12.
Blood ; 120(19): 4006-17, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22932805

ABSTRACT

The coding single nucleotide polymorphism GFI136N in the human gene growth factor independence 1 (GFI1) is present in 3%-7% of whites and increases the risk for acute myeloid leukemia (AML) by 60%. We show here that GFI136N, in contrast to GFI136S, lacks the ability to bind to the Gfi1 target gene that encodes the leukemia-associated transcription factor Hoxa9 and fails to initiate histone modifications that regulate HoxA9 expression. Consistent with this, AML patients heterozygous for the GFI136N variant show increased HOXA9 expression compared with normal controls. Using ChipSeq, we demonstrate that GFI136N specific epigenetic changes are also present in other genes involved in the development of AML. Moreover, granulomonocytic progenitors, a bone marrow subset from which AML can arise in humans and mice, show a proliferative expansion in the presence of the GFI136N variant. In addition, granulomonocytic progenitors carrying the GFI136N variant allele have altered gene expression patterns and differ in their ability to grow after transplantation. Finally, GFI136N can accelerate a K-RAS driven fatal myeloproliferative disease in mice. Our data suggest that the presence of a GFI136N variant allele induces a preleukemic state in myeloid precursors by deregulating the expression of Hoxa9 and other AML-related genes.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Homeodomain Proteins/genetics , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , Animals , Cluster Analysis , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Hematopoiesis/genetics , Histones/metabolism , Humans , Mice , Mice, Transgenic , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/mortality , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism
13.
J Exp Med ; 209(1): 187-99, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22201127

ABSTRACT

Precise regulation of Rag (recombination-activating gene) expression is crucial to prevent genomic instability caused by the generation of Rag-mediated DNA breaks. Although mechanisms of Rag activation have been well characterized, the mechanism by which Rag expression is down-regulated in early B cell development has not been fully elucidated. Using a complementary DNA library screen, we identified the transcriptional repressor Gfi1b as negative regulator of the Rag locus. Expression of Gfi1b causes repression of Rag1 and Rag2 in cell lines and primary mouse cells. Conversely, Gfi1b-deficient cell lines exhibit increased Rag expression, double-strand breaks and recombination, and cell cycle defects. In primary cells, transcription of Gfi1b inversely correlates with Rag transcription, and simultaneous inactivation of Gfi1 and Gfi1b leads to an increase in Rag transcription early in B cell development. In addition, deletion of Gfi1 and Gfi1b in vivo results in a severe block in B cell development. Gfi1b orchestrates Rag repression via a dual mechanism. Direct binding of Gfi1b to a site 5' of the B cell-specific Erag enhancer results in epigenetic changes in the Rag locus, whereas indirect inhibition is achieved through repression of the trans-activator Foxo1. Together, our experiments show that Gfi family members are essential for normal B cell development and play an important role in modulating expression of the V(D)J recombinase.


Subject(s)
Forkhead Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1 , Gene Deletion , Gene Expression Regulation , Gene Library , Gene Targeting , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Recombination, Genetic , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
14.
J Immunol ; 187(6): 2982-92, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841135

ABSTRACT

Miz-1 is a Broad-complex, Tramtrack and Bric-à-brac/pox virus zinc finger domain (BTB/POZ)-containing protein expressed in lymphoid precursors that can activate or repress transcription. We report in this article that mice expressing a nonfunctional Miz-1 protein lacking the BTB/POZ domain (Miz-1(ΔPOZ)) have a severe differentiation block at the pre-T cell "ß-selection" checkpoint, evident by a drastic reduction of CD4(-)CD8(-) double-negative-3 (DN3) and DN4 cell numbers. T cell-specific genes including Rag-1, Rag-2, CD3ε, pTα, and TCRß are expressed in Miz-1-deficient cells and V(D)J recombination is intact, but few DN3/DN4 cells express a surface pre-TCR. Miz-1-deficient DN3 cells are highly apoptotic and do not divide, which is consistent with enhanced expression of p53 target genes such as Cdkn1a, PUMA, and Noxa. However, neither coexpression of the antiapoptotic protein Bcl2 nor the deletion of p21(CIP1) nor the combination of both relieved Miz-1-deficient DN3/DN4 cells from their differentiation block. Only the coexpression of rearranged TCRαß and Bcl2 fully rescued Miz-1-deficient DN3/DN4 cell numbers and enabled them to differentiate into DN4TCRß(+) and double-positive cells. We propose that Miz-1 is a critical factor for the ß-selection checkpoint and is required for both the regulation of p53 target genes and proper expression of the pre-TCR to support the proliferative burst of DN3 cells during T cell development.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , Genes, T-Cell Receptor beta , Nuclear Proteins/immunology , Protein Inhibitors of Activated STAT/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , Animals , Cell Cycle , Cell Separation , Flow Cytometry , Gene Expression , Immunoblotting , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Protein Inhibitors of Activated STAT/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases
15.
Blood ; 117(12): 3370-81, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21258009

ABSTRACT

T cells originate from early T lineage precursors that have entered the thymus and differentiate through well-defined steps. Mice deficient for the BTB/POZ domain of zinc finger protein-1 (Miz-1) almost entirely lack early T lineage precursors and have a CD4(-)CD8(-) to CD4(+)CD8(+) block causing a strong reduction in thymic cellularity. Miz-1(ΔPOZ) pro-T cells cannot differentiate in vitro and are unable to relay signals from the interleukin-7R (IL-7R). Both STAT5 phosphorylation and Bcl-2 up-regulation are perturbed. The high expression levels of SOCS1 found in Miz-1(ΔPOZ) cells probably cause these alterations. Moreover, Miz-1 can bind to the SOCS1 promoter, suggesting that Miz-1 deficiency causes a deregulation of SOCS1. Transgenic overexpression of Bcl-2 or inhibition of SOCS1 restored pro-T cell numbers and their ability to differentiate, supporting the hypothesis that Miz-1 is required for the regulation of the IL-7/IL-7R/STAT5/Bcl-2 signaling pathway by monitoring the expression levels of SOCS1.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Kruppel-Like Transcription Factors/physiology , Lymphoid Progenitor Cells/physiology , Receptors, Interleukin-7/physiology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Gene Expression Regulation , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Lymphoid Progenitor Cells/immunology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Structure, Tertiary , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/physiology , Zinc Fingers/genetics
16.
Blood ; 116(24): 5149-61, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20826720

ABSTRACT

Donor-matched transplantation of hematopoietic stem cells (HSCs) is widely used to treat hematologic malignancies but is associated with high mortality. The expansion of HSC numbers and their mobilization into the bloodstream could significantly improve therapy. We report here that adult mice conditionally deficient for the transcription Growth factor independence 1b (Gfi1b) show a significant expansion of functional HSCs in the bone marrow and blood. Despite this expansion, Gfi1b(ko/ko) HSCs retain their ability to self-renew and to initiate multilineage differentiation but are no longer quiescent and contain elevated levels of reactive oxygen species. Treatment of Gfi1b(ko/ko) mice with N-acetyl-cystein significantly reduced HSC numbers indicating that increased reactive oxygen species levels are at least partially responsible for the expansion of Gfi1b-deficient HSCs. Moreover, Gfi1b(-/-) HSCs show decreased expression of CXCR4 and Vascular cell adhesion protein-1, which are required to retain dormant HSCs in the endosteal niche, suggesting that Gfi1b regulates HSC dormancy and pool size without affecting their function. Finally, the additional deletion of the related Gfi1 gene in Gfi1b(ko/ko) HSCs is incompatible with the maintenance of HSCs, suggesting that Gfi1b and Gfi1 have partially overlapping functions but that at least one Gfi gene is essential for the generation of HSCs.


Subject(s)
Cell Movement , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Acetylcysteine/pharmacology , Amine Oxidase (Copper-Containing)/biosynthesis , Animals , Cell Adhesion Molecules/biosynthesis , DNA-Binding Proteins/physiology , Homeostasis , Mice , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Reactive Oxygen Species , Receptors, CXCR4/biosynthesis , Repressor Proteins/deficiency , Transcription Factors/physiology
17.
Mol Cell Biol ; 30(16): 3929-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20547752

ABSTRACT

Endotoxin (bacterial lipopolysaccharide [LPS]) causes fatal septic shock via the Toll-like receptor 4 (TLR-4) protein present on innate immunity effector cells, which activates nuclear factor kappa B (NF-kappaB), inducing proinflammatory cytokines, including tumor necrosis factor alpha (TNF-alpha). An early step in this process involves nuclear sequestration of the p65-RelA NF-kappaB subunit, enabling transcriptional activation of target inflammatory cytokine genes. Here, we analyzed the role of the nuclear zinc finger protein Gfi1 in the TLR response using primary bone marrow-derived macrophages. We show that upon LPS stimulation, expression of Gfi1 is induced with kinetics similar to those of nuclear translocation of p65 and that Gfi1 interacts with p65 and inhibits p65-mediated transcriptional transactivation by interfering with p65 binding to target gene promoter DNA. Gfi1-deficient macrophages show abnormally high mRNA levels of the TNF-alpha gene and many other p65 target genes and a higher rate of TNF promoter occupancy by p65 than wild-type cells after LPS stimulation, suggesting that Gfi1 functions as an antagonist of NF-kappaB activity at the level of promoter binding. Our findings identify a new function of Gfi1 as a general negative regulator of the endotoxin-initiated innate immune responses, including septic shock and possibly other severe inflammatory diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Inflammation/immunology , Inflammation/metabolism , Toll-Like Receptors/metabolism , Transcription Factor RelA/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , DNA/genetics , DNA/metabolism , DNA Primers/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Humans , Immunity, Innate/drug effects , Inflammation/etiology , Lipopolysaccharides/toxicity , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Shock, Septic/etiology , Shock, Septic/immunology , Shock, Septic/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Zinc Fingers
18.
Int J Hematol ; 89(4): 422-430, 2009 May.
Article in English | MEDLINE | ID: mdl-19360458

ABSTRACT

Gfi1b is a transcriptional repressor that is essential for erythroid cells and megakaryocytes, but is also expressed in hematopoietic stem cells and early myeloid progenitors. The chromosomal localization of the Gfi1b gene at 9q34 and its functional homology with the proto-oncogene Gfi1 were suggestive for a role of Gfi1b in malignant transformation and myeloid leukemia. We show here that the expression of Gfi1b is strongly elevated in CML and AML patients compared to normal healthy controls and that imatinib, a drug widely used to treat CML, further enhances Gfi1b expression in patients even after remission. Our data suggest that Gfi1b may be an important factor to establish or maintain myeloid leukemia and myeloproliferative diseases and that, high expression levels of Gfi1b might be associated with the emergence of Philadelphia chromosome negative myeloid malignancies after imatinib withdrawal or after the development of imatinib resistance.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation, Neoplastic/genetics , Leukemia/genetics , Leukemia/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Acute Disease , Animals , Antineoplastic Agents/therapeutic use , Benzamides , COS Cells , Chlorocebus aethiops , Chronic Disease , Humans , Imatinib Mesylate , Leukemia/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Pyrimidines/therapeutic use , RNA, Messenger/genetics , Zinc Finger Protein GLI1
19.
Blood ; 109(6): 2356-64, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17095621

ABSTRACT

Gfi1b and Gfi1 are 37- and 55-kDa transcriptional repressors that share common features such as a 20-amino acid (aa) N-terminal SNAG domain, a nonconserved intermediary domain, and 6 highly conserved C-terminal zinc fingers. Both gene loci are under autoregulatory and cross-regulatory feedback control. We have generated a reporter mouse strain by inserting the cDNA for green fluorescent protein (GFP) into the Gfi1b gene locus which allowed us to follow Gfi1b expression during hematopoiesis and lymphopoiesis by measuring green fluorescence. We found highly dynamic expression patterns of Gfi1b in erythroid cells, megakaryocytes, and their progenitor cells (MEPS) where Gfi1 is not detected. Vice versa, Gfi1b could not be found in granulocytes, activated macrophages, or their granulomonocytic precursors (GMPs) or in mature naive or activated lymphocytes where Gfi1 is expressed, suggesting a complementary regulation of both loci during hematopoiesis. However, Gfi1b was found to be up-regulated in early stages of B-cell and in a subset of early T-cell development, where Gfi1 is also present, suggesting that cross-regulation of both loci exists but is cell-type specific.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression , Green Fluorescent Proteins/metabolism , Hematopoiesis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Aging/physiology , Animals , Biomarkers , Cell Lineage , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Green Fluorescent Proteins/genetics , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Stem Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
20.
EMBO J ; 25(11): 2409-19, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16688220

ABSTRACT

Gfi1b is a 37 kDa nuclear protein with six C2H2 zinc-finger domains that can silence transcription upon binding to specific target gene promoters. Here we show by using a chromatin immunoprecipitation and cloning protocol that Gfi1b also binds to gamma-satellite sequences that mainly occur in pericentric heterochromatin. Immuno-FISH experiments demonstrated that Gfi1b is localized at foci of pericentric heterochromatin identified by DAPI staining. Elevated levels of Gfi1b correlated with increased histone H3 lysine 9 dimethylation at sites neighboring gamma-satellite sequences but also at Gfi1b target gene promoters. In Gfi1b-deficient cells, however, a decrease of histone H3 lysine 9 trimethylation and a loss of heterochromatic structures was observed. Strikingly, we found that Gfi1b binds to both SUV39H1 and G9A histone methyl transferases, which provides a direct link between histone methylation and Gfi1b at heterochromatic and euchromatic sites. We propose that Gfi1b functions in heterochromatin formation and silencing of euchromatic transcription by recruiting histone methyl transferases to either gamma-satellite sequences or specific target gene promoters.


Subject(s)
DNA, Satellite/genetics , Heterochromatin/genetics , Histones/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , DNA, Satellite/metabolism , Gene Silencing , Heterochromatin/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Ikaros Transcription Factor/metabolism , In Situ Hybridization, Fluorescence , Methylation , Methyltransferases/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Protein Binding , Protein Methyltransferases , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Spleen/cytology , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...