Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Inflamm Regen ; 42(1): 6, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35227325

ABSTRACT

BACKGROUND: The expression of FZD5 distinguishes immature human mesenchymal stem/stromal cells (MSC) in cultures, and the function of FZD5 is crucial for maintaining the proliferation and multilineage differentiation capacity of human MSC. We herein investigated whether Fzd5 expression also marks undifferentiated MSC in animals. METHODS: We generated a transgenic mouse strain (Fzd5-CreERT-tFP635) that expresses CreERT and the fluorescent protein, TurboFP635 (tFP635), under the transcriptional control of the Fzd5 gene using the BAC transgenic technique, and identified cells expressing tFP635 by flow cytometry. We also conducted lineage tracing with this strain. RESULTS: In the bone marrow of transgenic mice, tFP635 was preferentially expressed in MSC, Leptin receptor-expressing MSC (LepR+MSCs), and some Pdgfrα+ Sca1+ MSC (PαS). Inducible lineage tracing using the Fzd5-CreERT-tFP635; CAG-CAT-EGFP strain at the adult stage showed that Fzd5-expressing cells and their descendants labeled with GFP were progressively dominant in LepR+MSC and PαS, and GFP+ cells persisted for 1 year after the activation of CreERT. Adipocyte progenitor cells (APCs), osteoblast progenitor cells (OPCs), and Cd51+ stromal cells were also labeled with GFP. CONCLUSIONS: Our transgenic mouse marks two different types of MSC, LepR+MSC and PαS.

2.
Curr Opin Hematol ; 27(4): 248-253, 2020 07.
Article in English | MEDLINE | ID: mdl-32398456

ABSTRACT

PURPOSE OF REVIEW: Even though an increasing amount of sequencing data on the leukemia genome has highlighted a tumor-suppressive function for plant homeodomain finger protein 6 (PHF6), its role in the hematopoietic system remained elusive until recently. The purpose of this review is to describe the role of PHF6 in normal hematopoiesis and leukemogenesis based on recent findings from knockout mouse models. RECENT FINDINGS: In a mouse model, the loss of Phf6 enhanced the bone marrow repopulating capacity of hematopoietic stem cells (HSCs) during serial transplantations without transforming hematopoietic cells, whereas donor mice, which lacked Phf6 expression in the hematopoietic system, did not show any apparent phenotypes in the steady-state. Mechanistically, Phf6 activates effectors in the tumor necrosis factor α (Tnfα) pathway. Therefore, a Phf6 deficiency attenuates the expression of the effectors and confers resistance against Tnfα-mediated growth inhibition to HSCs. Moreover, the loss of Phf6 promoted the development of leukemia induced by aberrant TLX3 expression or an active NOTCH mutation. SUMMARY: Phf6 restricts the self-renewal of HSCs by governing the Tnfα pathway. Phf6 fulfills a tumor-suppressive function, and its loss synergizes with leukemic lesions to promote the onset of hematological malignancies.


Subject(s)
Carcinogenesis/metabolism , Hematopoiesis , Leukemia/metabolism , Neoplasm Proteins/metabolism , Repressor Proteins/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia/genetics , Leukemia/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics
3.
Blood ; 134(14): 1154-1158, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31434704

ABSTRACT

KDM4/JMJD2 are H3K9- and H3K36-specific demethylases, which are considered promising therapeutic targets for the treatment of acute myeloid leukemia (AML) harboring MLL translocations. Here, we investigate the long-term effects of depleting KDM4 activity on normal hematopoiesis to probe potential side effects of continuous inhibition of these enzymes. Utilizing conditional Kdm4a/Kdm4b/Kdm4c triple-knockout mice, we show that KDM4 activity is required for hematopoietic stem cell (HSC) maintenance in vivo. The knockout of the KDM4 demethylases leads to accumulation of H3K9me3 on transcription start sites and the corresponding downregulation of expression of several genes in HSCs. We show that 2 of these genes, Taf1b and Nom1, are essential for the maintenance of hematopoietic cells. Taken together, our results show that the KDM4 demethylases are required for the expression of genes essential for the long-term maintenance of normal hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Histone Demethylases/genetics , Animals , Cell Survival , Cells, Cultured , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Initiation Site
4.
Exp Hematol ; 76: 24-37, 2019 08.
Article in English | MEDLINE | ID: mdl-31408689

ABSTRACT

The polycomb group protein Bmi1 maintains hematopoietic stem cell (HSC) functions. We previously reported that Bmi1-deficient mice exhibited progressive fatty changes in bone marrow (BM). A large portion of HSCs reside in the perivascular niche created partly by endothelial cells and leptin receptor+ (LepR+) BM stromal cells. To clarify how Bmi1 regulates the HSC niche, we specifically deleted Bmi1 in LepR+ cells in mice. The Bmi1 deletion promoted the adipogenic differentiation of LepR+ stromal cells and caused progressive fatty changes in the BM of limb bones with age, resulting in reductions in the numbers of HSCs and progenitors in BM and enhanced extramedullary hematopoiesis. This adipogenic change was also evident during BM regeneration after irradiation. Several adipogenic regulator genes appeared to be regulated by Bmi1. Our results indicate that Bmi1 keeps the adipogenic differentiation program repressed in BM stromal cells to maintain the integrity of the HSC niche.


Subject(s)
Adipogenesis/physiology , Hematopoietic Stem Cells/cytology , Polycomb Repressive Complex 1/physiology , Proto-Oncogene Proteins/physiology , Stem Cell Niche , Animals , Bone Marrow/pathology , Bone Marrow/physiology , Cell Line , Cell Self Renewal , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Polycomb Repressive Complex 1/deficiency , Proto-Oncogene Proteins/deficiency , Receptors, Leptin/analysis , Regeneration , Stromal Cells/chemistry , Stromal Cells/pathology
5.
Stem Cells Int ; 2019: 9789240, 2019.
Article in English | MEDLINE | ID: mdl-31065285

ABSTRACT

The "bivalent domain," a distinctive histone modification signature, is characterized by repressive trimethylation of histone H3 at lysine 27 (H3K27me3) and active trimethylation of histone H3 at lysine 4 (H3K4me3) marks. Maintenance and dynamic resolution of these histone marks play important roles in regulating differentiation processes in various stem cell systems. However, little is known regarding their roles in hepatic stem/progenitor cells. In the present study, we conducted the chromatin immunoprecipitation (ChIP) assay followed by high-throughput DNA sequencing (ChIP-seq) analyses in purified delta-like 1 protein (Dlk+) hepatic stem/progenitor cells and successfully identified 562 genes exhibiting bivalent domains within 2 kb of the transcription start site. Gene ontology analysis revealed that these genes were enriched in developmental functions and differentiation processes. Microarray analyses indicated that many of these genes exhibited derepression after differentiation toward hepatocyte and cholangiocyte lineages. Among these, 72 genes, including Cdkn2a and Sox4, were significantly upregulated after differentiation toward hepatocyte or cholangiocyte lineages. Knockdown of Sox4 in Dlk+ cells suppressed colony propagation and resulted in increased numbers of albumin+/cytokeratin 7+ progenitor cells in colonies. These findings implicate that derepression of Sox4 expression is required to induce normal differentiation processes. In conclusion, combined ChIP-seq and microarray analyses successfully identified bivalent genes. Functional analyses of these genes will help elucidate the epigenetic machinery underlying the terminal differentiation of hepatic stem/progenitor cells.

6.
Cell Microbiol ; 21(8): e13033, 2019 08.
Article in English | MEDLINE | ID: mdl-31009148

ABSTRACT

Vibrio cholerae produced-Cholix toxin (Cholix) is a cytotoxin that ADP-ribosylates eukaryotic elongation factor 2, inhibiting protein synthesis, and inducing apoptosis. Here, we identified prohibitin (PHB) 1 and 2 as novel Cholix-interacting membrane proteins in immortalised human hepatocytes and HepG2 cells by Cholix immunoprecipitation assays. The expression level of PHB1 was decreased by Cholix after a 12hr incubation. Cholix-induced poly (ADP-ribose) polymerase (PARP) cleavage was significantly enhanced in PHB (PHB1 or PHB2) knockdown cells. In contrast, transiently overexpressed PHB in hepatocytes attenuated Cholix-induced Bax/Bak conformational changes and PARP cleavage. In addition, Cholix-induced reactive oxygen species production and accumulation of fragmented mitochondria were enhanced in PHB-knockdown cells. Furthermore, Cholix induced activation of Rho-associated coiled coil-containing protein kinase 1 (ROCK1), which was enhanced in PHB-knockdown cells, followed by actin filament depolymerisation and accumulation of tubulin in the blebbing cells. Inhibition of ROCK1 by siRNA or its inhibitor suppressed Cholix-induced PARP cleavage and reactive oxygen species generation. Our findings identify PHB as a new protein that interacts with Cholix and is involved in Cholix-induced mitochondrial dysfunction and cytoskeletal rearrangement by ROCK1 activation during apoptosis.


Subject(s)
ADP-Ribosylation Factors/chemistry , Bacterial Toxins/chemistry , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions/genetics , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Vibrio cholerae/genetics , ADP-Ribosylation , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/microbiology , Actin Cytoskeleton/ultrastructure , Amino Acid Sequence , Apoptosis/genetics , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Line, Transformed , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/microbiology , Hepatocytes/pathology , Humans , Mitochondria/metabolism , Mitochondria/microbiology , Mitochondria/ultrastructure , Prohibitins , Protein Binding , Protein Isoforms/deficiency , Protein Isoforms/genetics , Proteolysis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Repressor Proteins/deficiency , Signal Transduction , Tubulin/genetics , Tubulin/metabolism , Vibrio cholerae/metabolism , Vibrio cholerae/pathogenicity , Virulence , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
7.
Blood ; 133(23): 2495-2506, 2019 06 06.
Article in English | MEDLINE | ID: mdl-30917958

ABSTRACT

Recurrent inactivating mutations have been identified in the X-linked plant homeodomain finger protein 6 (PHF6) gene, encoding a chromatin-binding transcriptional regulator protein, in various hematological malignancies. However, the role of PHF6 in normal hematopoiesis and its tumor-suppressor function remain largely unknown. We herein generated mice carrying a floxed Phf6 allele and inactivated Phf6 in hematopoietic cells at various developmental stages. The Phf6 deletion in embryos augmented the capacity of hematopoietic stem cells (HSCs) to proliferate in cultures and reconstitute hematopoiesis in recipient mice. The Phf6 deletion in neonates and adults revealed that cycling HSCs readily acquired an advantage in competitive repopulation upon the Phf6 deletion, whereas dormant HSCs only did so after serial transplantations. Phf6-deficient HSCs maintained an enhanced repopulating capacity during serial transplantations; however, they did not induce any hematological malignancies. Mechanistically, Phf6 directly and indirectly activated downstream effectors in tumor necrosis factor α (TNFα) signaling. The Phf6 deletion repressed the expression of a set of genes associated with TNFα signaling, thereby conferring resistance against the TNFα-mediated growth inhibition on HSCs. Collectively, these results not only define Phf6 as a novel negative regulator of HSC self-renewal, implicating inactivating PHF6 mutations in the pathogenesis of hematological malignancies, but also indicate that a Phf6 deficiency alone is not sufficient to induce hematopoietic transformation.


Subject(s)
Cell Self Renewal , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Proliferation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Blood ; 132(23): 2470-2483, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30228234

ABSTRACT

BCOR, encoding BCL-6 corepressor (BCOR), is X-linked and targeted by somatic mutations in various hematological malignancies including myelodysplastic syndrome (MDS). We previously reported that mice lacking Bcor exon 4 (Bcor ΔE4/y ) in the hematopoietic compartment developed NOTCH-dependent acute T-cell lymphoblastic leukemia (T-ALL). Here, we analyzed mice lacking Bcor exons 9 and 10 (Bcor ΔE9-10/y ), which express a carboxyl-terminal truncated BCOR that fails to interact with core effector components of polycomb repressive complex 1.1. Bcor ΔE9-10/y mice developed lethal T-ALL in a similar manner to Bcor ΔE4/y mice, whereas Bcor ΔE9-10/y hematopoietic cells showed a growth advantage in the myeloid compartment that was further enhanced by the concurrent deletion of Tet2 Tet2 Δ/Δ Bcor ΔE9-10/y mice developed lethal MDS with progressive anemia and leukocytopenia, inefficient hematopoiesis, and the morphological dysplasia of blood cells. Tet2 Δ/Δ Bcor ΔE9-10/y MDS cells reproduced MDS or evolved into lethal MDS/myeloproliferative neoplasms in secondary recipients. Transcriptional profiling revealed the derepression of myeloid regulator genes of the Cebp family and Hoxa cluster genes in Bcor ΔE9-10/y progenitor cells and the activation of p53 target genes specifically in MDS erythroblasts where massive apoptosis occurred. Our results reveal a tumor suppressor function of BCOR in myeloid malignancies and highlight the impact of Bcor insufficiency on the initiation and progression of MDS.


Subject(s)
Amino Acid Sequence , Exons , Myelodysplastic Syndromes , Repressor Proteins , Sequence Deletion , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Oncotarget ; 8(13): 21315-21326, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423509

ABSTRACT

Histone H3 lysine 9 dimethylation (H3K9me2) is mainly regulated by the histone lysine methyltransferase G9a and is associated with the repression of transcription. However, both the role of G9a and the significance of H3K9me2 in hepatocellular carcinoma (HCC) cells remain unclear. In this study, we conducted loss-of-function assay of G9a using short-hairpin RNA and pharmacological interference. Knockdown of G9a reduced H3K9me2 levels and impaired both HCC cell growth and sphere formation. However, transforming growth factor ß1-induced epithelial mesenchymal transition (EMT) was not suppressed by G9a knockdown. Combined analyses of chromatin immunoprecipitation followed by sequencing and RNA-sequencing led to successful identification of 96 candidate epigenetic targets of G9a. Pharmacological inhibition of G9a by BIX-01294 resulted in both cell growth inhibition and induction of apoptosis in HCC cells. Intraperitoneal administration of BIX-01294 suppressed the growth of xenograft tumors generated by implantation of HCC cells in non-obese diabetic/severe combined immunodeficient mice. Immunohistochemical analyses revealed high levels of G9a and H3K9me2 in 36 (66.7%) and 35 (64.8%) primary HCC tissues, respectively. G9a expression levels were significantly positively correlated with H3K9me2 levels in tumor tissues. In contrast, in non-tumor tissues, G9a and H3K9me2 were only observed in biliary epithelial cells and periportal hepatocytes. In conclusion, G9a inhibition impairs anchorage-dependent and -independent cell growth, but not EMT in HCC cells. Our data indicate that pharmacological interference of G9a might be a novel epigenetic approach for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Liver Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Azepines/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/pathology , Chromatin Immunoprecipitation , DNA Methylation , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens , Histones/genetics , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Quinazolines/pharmacology , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
10.
Blood ; 128(5): 638-49, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27301860

ABSTRACT

Setdb1, also known as Eset, is a methyltransferase that catalyzes trimethylation of H3K9 (H3K9me3) and plays an essential role in the silencing of endogenous retroviral elements (ERVs) in the developing embryo and embryonic stem cells (ESCs). Its role in somatic stem cells, however, remains unclear because of the early death of Setdb1-deficient embryos. We demonstrate here that Setdb1 is the first H3K9 methyltransferase shown to be essential for the maintenance of hematopoietic stem and progenitor cells (HSPCs) in mice. The deletion of Setdb1 caused the rapid depletion of hematopoietic stem and progenitor cells (HSPCs), as well as leukemic stem cells. In contrast to ESCs, ERVs were largely repressed in Setdb1-deficient HSPCs. A list of nonhematopoietic genes was instead ectopically activated in HSPCs after reductions in H3K9me3 levels, including key gluconeogenic enzyme genes fructose-1,6-bisphosphatase 1 (Fbp1) and Fbp2 The ectopic activation of gluconeogenic enzymes antagonized glycolysis and impaired ATP production, resulting in a compromised repopulating capacity of HSPCs. Our results demonstrate that Setdb1 maintains HSPCs by restricting the ectopic activation of nonhematopoietic genes detrimental to their function and uncover that the gluconeogenic pathway is one of the critical targets of Setdb1 in HSPCs.


Subject(s)
Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Animals , Bone Marrow/pathology , Endogenous Retroviruses/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Gene Deletion , Gene Silencing , Gluconeogenesis/genetics , Homeostasis/genetics , Leukemia/genetics , Leukemia/pathology , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
11.
Genes Dev ; 30(11): 1278-88, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27257215

ABSTRACT

Acute myeloid leukemias (AMLs) with a rearrangement of the mixed-linage leukemia (MLL) gene are aggressive hematopoietic malignancies. Here, we explored the feasibility of using the H3K9- and H3K36-specific demethylases Jmjd2/Kdm4 as putative drug targets in MLL-AF9 translocated leukemia. Using Jmjd2a, Jmjd2b, and Jmjd2c conditional triple-knockout mice, we show that Jmjd2/Kdm4 activities are required for MLL-AF9 translocated AML in vivo and in vitro. We demonstrate that expression of the interleukin 3 receptor α (Il3ra also known as Cd123) subunit is dependent on Jmjd2/Kdm4 through a mechanism involving removal of H3K9me3 from the promoter of the Il3ra gene. Importantly, ectopic expression of Il3ra in Jmjd2/Kdm4 knockout cells alleviates the requirement of Jmjd2/Kdm4 for the survival of AML cells, showing that Il3ra is a critical downstream target of Jmjd2/Kdm4 in leukemia. These results suggest that the JMJD2/KDM4 proteins are promising drug targets for the treatment of AML.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/physiopathology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Methylation , Mice , Mice, Knockout , Protein Binding , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
12.
Exp Hematol ; 44(4): 282-96.e3, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26773568

ABSTRACT

Fetal liver hematopoietic stem cells (HSCs) seed bone marrow (BM) and undergo reprograming into adult-type HSCs that are largely quiescent and restricted in their self-renewal activity. Here we report that in the absence of the polycomb-group gene Ezh2, a cohort of fetal-specific genes, including let-7 target genes, were activated in BM hematopoietic stem/progenitor cells (HSPCs), leading to acquisition of fetal phenotypes by BM HSPCs, such as enhanced self-renewal activity and production of fetal-type lymphocytes. The Lin28b/let-7 pathway determines developmentally timed changes in HSPC programs. Of note, many of the fetal-specific let-7 target genes, including Lin28, appear to be transcriptionally repressed by Ezh2-mediated H3K27me3 in BM HSPCs, and Ezh2 loss results in their ectopic expression, particularly in hematologic malignancies that develop in the absence of Ezh2. These findings suggest that Ezh2 cooperates with let-7 microRNAs in silencing the fetal gene signature in BM HSPCs and restricts their transformation.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Polycomb Repressive Complex 2/metabolism , RNA-Binding Proteins/genetics , Animals , Computational Biology/methods , Disease Progression , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Silencing , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Lymphopoiesis/genetics , Mice , Mice, Transgenic , Models, Biological , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/genetics , RNA Interference , Transcriptome
13.
PLoS One ; 10(11): e0142806, 2015.
Article in English | MEDLINE | ID: mdl-26571505

ABSTRACT

Alterations in chromatin structure caused by deregulated epigenetic mechanisms collaborate with underlying genetic lesions to promote cancer. SMARCA4/BRG1, a core component of the SWI/SNF ATP-dependent chromatin-remodelling complex, has been implicated by its mutational spectrum as exerting a tumour-suppressor function in many solid tumours; recently however, it has been reported to sustain leukaemogenic transformation in MLL-rearranged leukaemia in mice. Here we further explore the role of SMARCA4 and the two SWI/SNF subunits SMARCD2/BAF60B and DPF2/BAF45D in leukaemia. We observed the selective requirement for these proteins for leukaemic cell expansion and self-renewal in-vitro as well as in leukaemia. Gene expression profiling in human cells of each of these three factors suggests that they have overlapping functions in leukaemia. The gene expression changes induced by loss of the three proteins demonstrate that they are required for the expression of haematopoietic stem cell associated genes but in contrast to previous results obtained in mouse cells, the three proteins are not required for the expression of c-MYC regulated genes.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Leukemia/pathology , Muscle Proteins/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Gene Rearrangement , Leukemia/genetics , Mice , Myeloid Cells/pathology , Protein Subunits/metabolism , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic
14.
Oncotarget ; 6(7): 5102-17, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25671302

ABSTRACT

FUSE-binding protein (FBP)-interacting repressor (FIR) is a c-myc transcriptional suppressor. A splice variant of FIR that lacks exon 2 in the transcriptional repressor domain (FIRΔexon2) upregulates c-myc transcription by inactivating wild-type FIR. The ratio of FIRΔexon2/FIR mRNA was increased in human colorectal cancer and hepatocellular carcinoma tissues. Because FIRΔexon2 is considered to be a dominant negative regulator of FIR, FIR heterozygous knockout (FIR⁺/⁻) C57BL6 mice were generated. FIR complete knockout (FIR⁻/⁻) was embryonic lethal before E9.5; therefore, it is essential for embryogenesis. This strongly suggests that insufficiency of FIR is crucial for carcinogenesis. FIR⁺/⁻ mice exhibited prominent c-myc mRNA upregulation, particularly in the peripheral blood (PB), without any significant pathogenic phenotype. Furthermore, elevated FIRΔexon2/FIR mRNA expression was detected in human leukemia samples and cell lines. Because the single knockout of TP53 generates thymic lymphoma, FIR⁺/⁻TP53⁻/⁻ generated T-cell type acute lymphocytic/lymphoblastic leukemia (T-ALL) with increased organ or bone marrow invasion with poor prognosis. RNA-sequencing analysis of sorted thymic lymphoma cells revealed that the Notch signaling pathway was activated significantly in FIR⁺/⁻TP53⁻/⁻ compared with that in FIR⁺/⁺TP53⁻/⁻ mice. Notch1 mRNA expression in sorted thymic lymphoma cells was confirmed using qRT-PCR. In addition, flow cytometry revealed that c-myc mRNA was negatively correlated with FIR but positively correlated with Notch1 in sorted T-ALL/thymic lymphoma cells. Moreover, the knockdown of TP53 or c-myc using siRNA decreased Notch1 expression in cancer cells. In addition, an adenovirus vector encoding FIRΔexon2 cDNA increased bleomycin-induced DNA damage. Taken together, these data suggest that the altered expression of FIRΔexon2 increased Notch1 at least partially by activating c-Myc via a TP53-independent pathway. In conclusion, the alternative splicing of FIR, which generates FIRΔexon2, may contribute to both colorectal carcinogenesis and leukemogenesis.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA-Binding Proteins/genetics , Receptor, Notch1/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Adult , Alternative Splicing , Animals , Disease Progression , Female , Haploinsufficiency , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing Factors , RNA-Binding Proteins/metabolism , Receptor, Notch1/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
15.
Nat Commun ; 5: 5872, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25519988

ABSTRACT

During T-cell development, Cd8 expression is controlled via dynamic regulation of its cis-regulatory enhancer elements. Insufficiency of enhancer activity causes variegated Cd8 expression in CD4(+)CD8(+) double-positive (DP) thymocytes. Brd1 is a subunit of the Hbo1 histone acetyltransferase (HAT) complex responsible for acetylation of histone H3 at lysine 14 (H3K14). Here we show that deletion of Brd1 in haematopoietic progenitors causes variegated expression of Cd8, resulting in the appearance of CD4(+)CD8(-)TCRß(-/low) thymocytes indistinguishable from DP thymocytes in their properties. Biochemical analysis confirms that Brd1 forms a HAT complex with Hbo1 in thymocytes. ChIP analysis demonstrates that Brd1 localizes at the known enhancers in the Cd8 genes and is responsible for acetylation at H3K14. These findings indicate that the Brd1-mediated HAT activity is crucial for efficient activation of Cd8 expression via acetylation at H3K14, which serves as an epigenetic mark that promotes the recruitment of transcription machinery to the Cd8 enhancers.


Subject(s)
CD8 Antigens/immunology , Epigenesis, Genetic , Histone Acetyltransferases/immunology , Protein Processing, Post-Translational , Thymocytes/immunology , Acetylation , Animals , CD8 Antigens/genetics , Cell Differentiation , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histones/genetics , Histones/immunology , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Thymocytes/cytology
16.
Stem Cell Reports ; 2(2): 145-52, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24527388

ABSTRACT

TIF1ß is a transcriptional corepressor that recruits repressive chromatin modifiers to target genes. Its biological function and physiological targets in somatic stem cells remain largely unknown. Here, we show that TIF1ß is essential for the maintenance of hematopoietic stem cells (HSCs). Deletion of Tif1b in mice induced active cycling and apoptosis of HSCs and promoted egression of HSCs from the bone marrow, leading to rapid depletion of HSCs. Strikingly, Tif1b-deficient HSCs showed a strong trend of ectopic expression of nonhematopoietic genes. Levels of heterochromatin protein 1 (HP1α, ß and γ) proteins, which form a complex with TIF1ß, were significantly reduced in the absence of TIF1ß and depletion of HP1 recapitulated a part of the phenotypes of Tif1b-deficient HSCs. These results demonstrate that the TIF1ß-HP1 system functions as a critical repressive machinery that targets genes not normally activated in the hematopoietic compartment, thereby maintaining the transcriptional signature specific to HSCs.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Hematopoietic Stem Cells/metabolism , Transcription, Genetic , Animals , Apoptosis/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Cycle/genetics , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation , Gene Knockout Techniques , Gene Targeting , Hematopoiesis/genetics , Liver/cytology , Liver/metabolism , Mice , Phenotype , Protein Binding , RNA Interference
17.
Blood ; 123(12): 1870-82, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24501218

ABSTRACT

Epigenetic regulatory mechanisms are implicated in the pathogenesis of acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL). Recent progress suggests that proteins involved in epigenetic control are amenable to drug intervention, but little is known about the cancer-specific dependency on epigenetic regulators for cell survival and proliferation. We used a mouse model of human AML induced by the MLL-AF9 fusion oncogene and an epigenetic short hairpin RNA (shRNA) library to screen for novel potential drug targets. As a counter-screen for general toxicity of shRNAs, we used normal mouse bone marrow cells. One of the best candidate drug targets identified in these screens was Jmjd1c. Depletion of Jmjd1c impairs growth and colony formation of mouse MLL-AF9 cells in vitro as well as establishment of leukemia after transplantation. Depletion of JMJD1C impairs expansion and colony formation of human leukemic cell lines, with the strongest effect observed in the MLL-rearranged ALL cell line SEM. In both mouse and human leukemic cells, the growth defect upon JMJD1C depletion appears to be primarily due to increased apoptosis, which implicates JMJD1C as a potential therapeutic target in leukemia.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Myeloid, Acute/genetics , Oxidoreductases, N-Demethylating/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Epigenesis, Genetic , Gene Knockdown Techniques , Genes, myb , Genes, myc , Histone-Lysine N-Methyltransferase/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Oxidoreductases, N-Demethylating/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , Tumor Stem Cell Assay
18.
PLoS One ; 9(1): e84807, 2014.
Article in English | MEDLINE | ID: mdl-24454751

ABSTRACT

Tumor-initiating cells (TICs) play a central role in tumor development, metastasis, and recurrence. In the present study, we investigated the effect of disulfiram (DSF), an inhibitor of aldehyde dehydrogenase, toward tumor-initiating hepatocellular carcinoma (HCC) cells. DSF treatment suppressed the anchorage-independent sphere formation of both HCC cells. Flow cytometric analyses showed that DSF but not 5-fluorouracil (5-FU) drastically reduces the number of tumor-initiating HCC cells. The sphere formation assays of epithelial cell adhesion molecule (EpCAM)(+) HCC cells co-treated with p38-specific inhibitor revealed that DSF suppresses self-renewal capability mainly through the activation of reactive oxygen species (ROS)-p38 MAPK pathway. Microarray experiments also revealed the enrichment of the gene set involved in p38 MAPK signaling in EpCAM(+) cells treated with DSF but not 5-FU. In addition, DSF appeared to downregulate Glypican 3 (GPC3) in a manner independent of ROS-p38 MAPK pathway. GPC3 was co-expressed with EpCAM in HCC cell lines and primary HCC cells and GPC3-knockdown reduced the number of EpCAM(+) cells by compromising their self-renewal capability and inducing the apoptosis. These results indicate that DSF impaired the tumorigenicity of tumor-initiating HCC cells through activation of ROS-p38 pathway and in part through the downregulation of GPC3. DSF might be a promising therapeutic agent for the eradication of tumor-initiating HCC cells.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Disulfiram/pharmacology , Liver Neoplasms/enzymology , MAP Kinase Signaling System/drug effects , Neoplastic Stem Cells/pathology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Antigens, Neoplasm/metabolism , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/metabolism , Cell Count , Cell Line, Tumor , Cell Proliferation/drug effects , Disulfiram/therapeutic use , Enzyme Activation/drug effects , Epithelial Cell Adhesion Molecule , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glypicans/genetics , Glypicans/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
19.
J Exp Med ; 210(12): 2627-39, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24218139

ABSTRACT

Polycomb group (PcG) proteins are essential regulators of hematopoietic stem cells. Recent extensive mutation analyses of the myeloid malignancies have revealed that inactivating somatic mutations in PcG genes such as EZH2 and ASXL1 occur frequently in patients with myelodysplastic disorders including myelodysplastic syndromes (MDSs) and MDS/myeloproliferative neoplasm (MPN) overlap disorders (MDS/MPN). In our patient cohort, EZH2 mutations were also found and often coincided with tet methylcytosine dioxygenase 2 (TET2) mutations. Consistent with these findings, deletion of Ezh2 alone was enough to induce MDS/MPN-like diseases in mice. Furthermore, concurrent depletion of Ezh2 and Tet2 established more advanced myelodysplasia and markedly accelerated the development of myelodysplastic disorders including both MDS and MDS/MPN. Comprehensive genome-wide analyses in hematopoietic progenitor cells revealed that upon deletion of Ezh2, key developmental regulator genes were kept transcriptionally repressed, suggesting compensation by Ezh1, whereas a cohort of oncogenic direct and indirect polycomb targets became derepressed. Our findings provide the first evidence of the tumor suppressor function of EZH2 in myeloid malignancies and highlight the cooperative effect of concurrent gene mutations in the pathogenesis of myelodysplastic disorders.


Subject(s)
DNA-Binding Proteins/genetics , Myelodysplastic Syndromes/etiology , Polycomb Repressive Complex 2/genetics , Proto-Oncogene Proteins/genetics , Animals , Cohort Studies , DNA-Binding Proteins/deficiency , Dioxygenases , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Genome-Wide Association Study , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Polycomb Repressive Complex 2/deficiency , Proto-Oncogene Proteins/deficiency , Tumor Suppressor Proteins/genetics
20.
Blood ; 121(3): 447-58, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23169777

ABSTRACT

To search for genes that promote hematopoietic development from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), we overexpressed several known hematopoietic regulator genes in hESC/iPSC-derived CD34(+)CD43(-) endothelial cells (ECs) enriched in hemogenic endothelium (HE). Among the genes tested, only Sox17, a gene encoding a transcription factor of the SOX family, promoted cell growth and supported expansion of CD34(+)CD43(+)CD45(-/low) cells expressing the HE marker VE-cadherin. SOX17 was expressed at high levels in CD34(+)CD43(-) ECs compared with low levels in CD34(+)CD43(+)CD45(-) pre-hematopoietic progenitor cells (pre-HPCs) and CD34(+)CD43(+)CD45(+) HPCs. Sox17-overexpressing cells formed semiadherent cell aggregates and generated few hematopoietic progenies. However, they retained hemogenic potential and gave rise to hematopoietic progenies on inactivation of Sox17. Global gene-expression analyses revealed that the CD34(+)CD43(+)CD45(-/low) cells expanded on overexpression of Sox17 are HE-like cells developmentally placed between ECs and pre-HPCs. Sox17 overexpression also reprogrammed both pre-HPCs and HPCs into HE-like cells. Genome-wide mapping of Sox17-binding sites revealed that Sox17 activates the transcription of key regulator genes for vasculogenesis, hematopoiesis, and erythrocyte differentiation directly. Depletion of SOX17 in CD34(+)CD43(-) ECs severely compromised their hemogenic activity. These findings suggest that SOX17 plays a key role in priming hemogenic potential in ECs, thereby regulating hematopoietic development from hESCs/iPSCs.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , SOXF Transcription Factors/genetics , SOXF Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/physiology , Fetal Blood/cytology , Fibroblasts/cytology , Hematopoiesis/genetics , Humans , Lentivirus/genetics , Mice , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Transduction, Genetic/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...