Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Proc Natl Acad Sci U S A ; 120(32): e2206860120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37523546

ABSTRACT

Mbtd1 (mbt domain containing 1) encodes a nuclear protein containing a zinc finger domain and four malignant brain tumor (MBT) repeats. We previously generated Mbtd1-deficient mice and found that MBTD1 is highly expressed in fetal hematopoietic stem cells (HSCs) and sustains the number and function of fetal HSCs. However, since Mbtd1-deficient mice die soon after birth possibly due to skeletal abnormalities, its role in adult hematopoiesis remains unclear. To address this issue, we generated Mbtd1 conditional knockout mice and analyzed adult hematopoietic tissues deficient in Mbtd1. We observed that the numbers of HSCs and progenitors increased and Mbtd1-deficient HSCs exhibited hyperactive cell cycle, resulting in a defective response to exogenous stresses. Mechanistically, we found that MBTD1 directly binds to the promoter region of FoxO3a, encoding a forkhead protein essential for HSC quiescence, and interacts with components of TIP60 chromatin remodeling complex and other proteins involved in HSC and other stem cell functions. Restoration of FOXO3a activity in Mbtd1-deficient HSCs in vivo rescued cell cycle and pool size abnormalities. These findings indicate that MBTD1 is a critical regulator for HSC pool size and function, mainly through the maintenance of cell cycle quiescence by FOXO3a.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Animals , Mice , Cell Cycle/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/metabolism
3.
Blood ; 137(7): 908-922, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33174606

ABSTRACT

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Subject(s)
Aging/genetics , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hematopoietic System/physiology , Histone Code/genetics , Histone Demethylases/physiology , Animals , Cellular Senescence/genetics , DNA Breaks, Double-Stranded , DNA Repair , Female , Genetic Predisposition to Disease , Hematopoiesis, Extramedullary , Histone Demethylases/deficiency , Histone Demethylases/genetics , Immune Reconstitution , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Male , Mice , Mice, Knockout , Moloney murine leukemia virus/physiology , Myeloid Cells/pathology , Radiation Chimera , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Virus Integration
4.
Clin Cancer Res ; 26(8): 2065-2079, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32047002

ABSTRACT

PURPOSE: Epigenetic deregulation is deeply implicated in the pathogenesis of bladder cancer. KDM6A (Lysine (K)-specific demethylase 6A) is a histone modifier frequently mutated in bladder cancer. However, the molecular mechanisms of how KDM6A deficiency contributes to bladder cancer development remains largely unknown. We hypothesized that clarification of the pathogenic mechanisms underlying KDM6A-mutated bladder cancer can help in designing new anticancer therapies. EXPERIMENTAL DESIGN: We generated mice lacking Kdm6a in the urothelium and crossed them with mice heterozygous for p53, whose mutation/deletion significantly overlaps with the KDM6A mutation in muscle-invasive bladder cancer (MIBC). In addition, BBN (N-butyl-N-(4-hydroxybutyl) nitrosamine), a cigarette smoke-like mutagen, was used as a tumor-promoting agent. Isolated urothelia were subjected to phenotypic, pathologic, molecular, and cellular analyses. The clinical relevance of our findings was further analyzed using genomic and clinical data of patients with MIBC. RESULTS: We found that Kdm6a deficiency activated cytokine and chemokine pathways, promoted M2 macrophage polarization, increased cancer stem cells and caused bladder cancer in cooperation with p53 haploinsufficiency. We also found that BBN treatment significantly enhanced the expression of proinflammatory molecules and accelerated disease development. Human bladder cancer samples with decreased KDM6A expression also showed activated proinflammatory pathways. Notably, dual inhibition of IL6 and chemokine (C-C motif) ligand 2, upregulated in response to Kdm6a deficiency, efficiently suppressed Kdm6a-deficient bladder cancer cell growth. CONCLUSIONS: Our findings provide insights into multistep carcinogenic processes of bladder cancer and suggest molecular targeted therapeutic approaches for patients with bladder cancer with KDM6A dysfunction.


Subject(s)
Carcinogenesis/pathology , Histone Demethylases/physiology , Inflammation/pathology , Macrophages/immunology , Tumor Suppressor Protein p53/physiology , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Databases, Genetic/statistics & numerical data , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Urothelium/immunology
5.
Blood ; 129(15): 2148-2160, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28209720

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a hematological malignancy characterized by uncontrolled proliferation of dysplastic myelomonocytes and frequent progression to acute myeloid leukemia (AML). We identified mutations in the Cbl gene, which encodes a negative regulator of cytokine signaling, in a subset of CMML patients. To investigate the contribution of mutant Cbl in CMML pathogenesis, we generated conditional knockin mice for Cbl that express wild-type Cbl in a steady state and inducibly express CblQ367P , a CMML-associated Cbl mutant. CblQ367P mice exhibited sustained proliferation of myelomonocytes, multilineage dysplasia, and splenomegaly, which are the hallmarks of CMML. The phosphatidylinositol 3-kinase (PI3K)-AKT and JAK-STAT pathways were constitutively activated in CblQ367P hematopoietic stem cells, which promoted cell cycle progression and enhanced chemokine-chemokine receptor activity. Gem, a gene encoding a GTPase that is upregulated by CblQ367P , enhanced hematopoietic stem cell activity and induced myeloid cell proliferation. In addition, Evi1, a gene encoding a transcription factor, was found to cooperate with CblQ367P and progress CMML to AML. Furthermore, targeted inhibition for the PI3K-AKT and JAK-STAT pathways efficiently suppressed the proliferative activity of CblQ367P -bearing CMML cells. Our findings provide insights into the molecular mechanisms underlying mutant Cbl-induced CMML and propose a possible molecular targeting therapy for mutant Cbl-carrying CMML patients.


Subject(s)
Cell Cycle , Hematopoietic Stem Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Mutation, Missense , Myelopoiesis , Proto-Oncogene Proteins c-cbl , Up-Regulation , Amino Acid Substitution , Animals , Gene Expression Regulation, Enzymologic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , Monocytes/metabolism , Monocytes/pathology , Monomeric GTP-Binding Proteins/biosynthesis , Monomeric GTP-Binding Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/biosynthesis , Proto-Oncogene Proteins c-cbl/genetics , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 113(37): 10370-5, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27578866

ABSTRACT

Polycomb repressive complex 2 (PRC2) catalyzes the monomethylation, dimethylation, and trimethylation of histone H3 Lys27 (H3K27) and acts as a central epigenetic regulator that marks the repressive chromatin domain. Embryonic ectoderm development (EED), an essential component of PRC2, interacts with trimethylated H3K27 (H3K27me3) through the aromatic cage structure composed of its three aromatic amino acids, Phe97, Trp364, and Tyr365. This interaction allosterically activates the histone methyltransferase activity of PRC2 and thereby propagates repressive histone marks. In this study, we report the analysis of knock-in mice harboring the myeloid disorder-associated EED Ile363Met (I363M) mutation, analogous to the EED aromatic cage mutants. The I363M homozygotes displayed a remarkable and preferential reduction of H3K27me3 and died at midgestation. The heterozygotes increased the clonogenic capacity and bone marrow repopulating activity of hematopoietic stem/progenitor cells (HSPCs) and were susceptible to leukemia. Lgals3, a PRC2 target gene encoding a multifunctional galactose-binding lectin, was derepressed in I363M heterozygotes, which enhanced the stemness of HSPCs. Thus, our work provides in vivo evidence that the structural integrity of EED to H3K27me3 propagation is critical, especially for embryonic development and hematopoietic homeostasis, and that its perturbation increases the predisposition to hematologic malignancies.


Subject(s)
Galectin 3/genetics , Leukemia/genetics , Polycomb Repressive Complex 2/chemistry , Animals , Embryonic Development/genetics , Epigenesis, Genetic/genetics , Galectin 3/chemistry , Genetic Predisposition to Disease , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Polycomb Repressive Complex 2/genetics
7.
Sci Rep ; 6: 29454, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27432459

ABSTRACT

Polycomb repressive complex 2 (PRC2) participates in transcriptional repression through methylation of histone H3K27. The WD-repeat protein embryonic ectoderm development (EED) is a non-catalytic but an essential component of PRC2 and its mutations were identified in hematopoietic malignancies. To clarify the role(s) of EED in adult hematopoiesis and leukemogenesis, we generated Eed conditional knockout (Eed(Δ/Δ)) mice. Eed(Δ/Δ) mice died in a short period with rapid decrease of hematopoietic cells. Hematopoietic stem/progenitor cells (HSPCs) were markedly decreased with impaired bone marrow (BM) repopulation ability. Cell cycle analysis of HSPCs demonstrated increased S-phase fraction coupled with suppressed G0/G1 entry. Genes encoding cell adhesion molecules are significantly enriched in Eed(Δ/Δ) HSPCs, and consistently, Eed(Δ/Δ) HSPCs exhibited increased attachment to a major extracellular matrix component, fibronectin. Thus, EED deficiency increases proliferation on one side but promotes quiescence possibly by enhanced adhesion to the hematopoietic niche on the other, and these conflicting events would lead to abnormal differentiation and functional defect of Eed(Δ/Δ) HSPCs. In addition, Eed haploinsufficiency induced hematopoietic dysplasia, and Eed heterozygous mice were susceptible to malignant transformation and developed leukemia in cooperation with Evi1 overexpression. Our results demonstrated differentiation stage-specific and dose-dependent roles of EED in normal hematopoiesis and leukemogenesis.


Subject(s)
Haploinsufficiency , Hematopoiesis , Leukemia/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/physiology , Animals , Antigens, CD34/metabolism , Cell Adhesion , Cell Cycle , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Extracellular Matrix/metabolism , Female , Fetal Blood/cytology , Fibronectins/chemistry , Fibronectins/metabolism , Flow Cytometry , Gene Transfer Techniques , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematopoietic Stem Cells/cytology , Heterozygote , Histones , Leukemia/genetics , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism
8.
Blood ; 125(22): 3437-46, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25872778

ABSTRACT

We previously reported that deficiency for Samd9L, which was cloned as a candidate gene for -7/7q- syndrome, accelerated leukemia cooperatively with enhanced expression of a histone demethylase: F-box and leucine-rich repeat protein 10 (Fbxl10, also known as Jhdm1b, Kdm2b, and Ndy1). To further investigate the role of Fbxl10 in leukemogenesis, we generated transgenic (Tg) mice that overexpress Fbxl10 in hematopoietic stem cells (HSCs). Interestingly, Fbxl10 Tg mice developed myeloid or B-lymphoid leukemia with complete penetrance. HSCs from the Tg mice exhibited an accelerated G0/G1-to-S transition with a normal G0 to G1 entry, resulting in pleiotropic progenitor cell expansion. Fbxl10 Tg HSCs displayed enhanced expression of neuron-specific gene family member 2 (Nsg2), and forced expression of Nsg2 in primary bone marrow cells resulted in expansion of immature cells. In addition, the genes involved in mitochondrial oxidative phosphorylation were markedly enriched in Fbxl10 Tg HSCs, coupled with increased cellular adenosine 5'-triphosphate levels. Moreover, chromatin immunoprecipitation followed by sequencing analysis demonstrated that Fbxl10 directly binds to the regulatory regions of Nsg2 and oxidative phosphorylation genes. These findings define Fbxl10 as a bona fide oncogene, whose deregulated expression contributes to the development of leukemia involving metabolic proliferative advantage and Nsg2-mediated impaired differentiation.


Subject(s)
Carrier Proteins/metabolism , F-Box Proteins/genetics , Hematopoietic Stem Cells/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia/genetics , Leukemia/metabolism , Nerve Tissue Proteins/metabolism , Animals , B-Lymphocytes/pathology , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , F-Box Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Nerve Tissue Proteins/genetics , Oncogenes , Up-Regulation/genetics
9.
PLoS One ; 9(1): e87425, 2014.
Article in English | MEDLINE | ID: mdl-24498102

ABSTRACT

A20 is a negative regulator of NF-κB, and mutational loss of A20 expression is involved in the pathogenesis of autoimmune diseases and B-cell lymphomas. To clarify the role of A20 in adult hematopoiesis, we generated conditional A20 knockout mice (A20(flox/flox) ) and crossed them with Mx-1Cre (MxCre (+)) and ERT2Cre (ERT2Cre (+)) transgenic mice in which Cre is inducibly activated by endogenous interferon and exogenous tamoxifen, respectively. A20(flox/flox) MxCre (+) (A20Mx) mice spontaneously exhibited myeloid proliferation, B cell apoptosis, and anemia with overproduction of pro-inflammatory cytokines. Bone marrow transplantation demonstrated that these changes were caused by hematopoietic cells. NF-κB was constitutively activated in A20Mx hematopoietic stem cells (HSCs), which caused enhanced cell cycle entry and impaired repopulating ability. Tamoxifen stimulation of A20(flox/flox) ERT2Cre (+) (A20ERT2) mice induced fulminant apoptosis and subsequent myeloproliferation, lymphocytopenia, and progressive anemia with excessive production of pro-inflammatory cytokines, as observed in A20Mx mice. These results demonstrate that A20 plays essential roles in the homeostasis of adult hematopoiesis by preventing apoptosis and inflammation. Our findings provide insights into the mechanism underlying A20 dysfunction and human diseases in which A20 expression is impaired.


Subject(s)
Apoptosis/physiology , Cell Proliferation , DNA-Binding Proteins/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cysteine Endopeptidases , Cytokines/metabolism , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Myeloid Cells/cytology , NF-kappa B/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases/genetics
10.
FEBS Lett ; 587(10): 1529-35, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23583447

ABSTRACT

CIZ1 is a nuclear protein involved in DNA replication and is also implicated in human diseases including cancers. To gain an insight into its function in vivo, we generated mice lacking Ciz1. Ciz1-deficient (Ciz1(-/-)) mice grew without any obvious abnormalities, and Ciz1(-/-) mouse embryonic fibroblasts (MEFs) did not show any defects in cell cycle status, cell growth, and DNA damage response. However, Ciz1(-/-) MEFs were sensitive to hydroxyurea-mediated replication stress and susceptible to oncogene-induced cellular transformation. In addition, Ciz1(-/-) mice developed various types of leukemias by retroviral insertional mutagenesis. These results indicate that CIZ1 functions as a tumor suppressor in vivo.


Subject(s)
Nuclear Proteins/physiology , Tumor Suppressor Proteins , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Fibroblasts/physiology , Mice , Mice, Knockout , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology
11.
BMC Immunol ; 13: 72, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23268649

ABSTRACT

BACKGROUND: CD72 is an inhibitory co-receptor expressed on B cells. We previously demonstrated significant association of the polymorphism of the CD72 gene with susceptibility to human systemic lupus erythematosus (SLE) in individuals carrying a SLE-susceptible FCGR2B genotype (FCGR2B-232Thr/Thr). The human CD72 locus generates a splicing isoform that lacks exon 8 (CD72Δex8) as well as full-length CD72 (CD72fl), and the CD72 polymorphism regulates exon 8 skipping. RESULTS: Here we demonstrated that individuals carrying the disease-protective CD72 genotype exhibit significantly lower serum immunoglobulin levels than do individuals carrying other CD72 genotypes (P < 0.05). Although expression level of CD72fl in the peripheral blood B cells was similar regardless of CD72 genotype, the protein level of CD72Δex8 was increased in individuals carrying the disease-protective CD72 genotype, suggesting a crucial role of CD72Δex8 in regulation of antibody production. By expressing these human CD72 isoforms in mouse cell lines, we further demonstrated that CD72Δex8 is accumulated in endoplasmic reticulum (ER) and fails to regulate BCR signaling whereas human CD72fl is efficiently transported to the cell surface and inhibits signaling through the B cell antigen receptor (BCR), as is the case for mouse CD72. CONCLUSION: Human CD72 polymorphism appears to regulate antibody production as well as susceptibility to SLE by regulating expression of ER-localizing CD72Δex8.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/immunology , Endoplasmic Reticulum/metabolism , Lupus Erythematosus, Systemic/immunology , Mutant Proteins/metabolism , Protein Isoforms/metabolism , Animals , Antibody Formation/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Cell Line , Genetic Predisposition to Disease , Humans , Immunoglobulins/blood , Mice , Mutant Proteins/genetics , Mutant Proteins/immunology , Polymorphism, Genetic , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Splicing , Protein Transport , Transgenes/genetics
12.
Proc Natl Acad Sci U S A ; 108(6): 2468-73, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21252303

ABSTRACT

To clarify the molecular pathways governing hematopoietic stem cell (HSC) development, we screened a fetal liver (FL) HSC cDNA library and identified a unique gene, hematopoietic expressed mammalian polycomb (hemp), encoding a protein with a zinc-finger domain and four malignant brain tumor (mbt) repeats. To investigate its biological role, we generated mice lacking Hemp (hemp(-/-)). Hemp(-/-) mice exhibited a variety of skeletal malformations and died soon after birth. In the FL, hemp was preferentially expressed in the HSC and early progenitor cell fractions, and analyses of fetal hematopoiesis revealed that the number of FL mononuclear cells, including HSCs, was reduced markedly in hemp(-/-) embryos, especially during early development. In addition, colony-forming and competitive repopulation assays demonstrated that the proliferative and reconstitution abilities of hemp(-/-) FL HSCs were significantly impaired. Microarray analysis revealed alterations in the expression levels of several genes implicated in hematopoietic development and differentiation in hemp(-/-) FL HSCs. These results demonstrate that Hemp, an mbt-containing protein, plays essential roles in HSC function and skeletal formation. It is also hypothesized that Hemp might be involved in certain congenital diseases, such as Klippel-Feil anomaly.


Subject(s)
Bone Development/physiology , Bone and Bones/embryology , Chromosomal Proteins, Non-Histone/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/physiology , Chromosomal Proteins, Non-Histone/genetics , Embryo, Mammalian/cytology , Gene Expression Profiling , Hematopoiesis/physiology , Klippel-Feil Syndrome/genetics , Klippel-Feil Syndrome/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Repressor Proteins/genetics
13.
J Immunol ; 185(12): 7330-9, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21057086

ABSTRACT

Naturally occurring regulatory T (Treg) cells play a central role in the maintenance of immune homeostasis and in restraining the development of spontaneous inflammatory responses. However, the underlying mechanisms of Treg homeostasis remain incompletely understood. Of particular note, the IL-2Rα (CD25) is crucial for the homeostasis of Treg cells and the prevention of lymphoproliferative autoimmune disease. In this paper, we report that the basic helix-loop-helix transcription factor Dec1 is involved in the homeostasis of Treg cells and plays a role in their survival or expansion after adoptive transfer to lymphopenic recipients. Hence, it is crucial for the suppression of effector T cell-mediated inflammatory responses. Enforced expression of Dec1 upregulates CD25 expression during thymocyte development and increases the number of Treg cells in the periphery. Dec1 binds the transcription factor Runx1 and colocalizes with Runx1 in Treg cells. Specifically, we demonstrate that in Treg cells the Dec1/Runx1 complex binds to regulatory elements present in the Il-2rα locus. Collectively, these data show how Dec1 mechanistically acts in Treg cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Homeodomain Proteins/immunology , Homeostasis/physiology , Interleukin-2 Receptor alpha Subunit/immunology , Up-Regulation/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Core Binding Factor Alpha 2 Subunit/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Transgenic , Protein Binding/genetics , Protein Binding/immunology , T-Lymphocytes, Regulatory
14.
Dev Biol ; 345(2): 226-36, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20647008

ABSTRACT

Although internal ribosome entry site (IRES)-mediated translation is considered important for proper cellular function, its precise biological role is not fully understood. Runx1 gene, which encodes a transcription factor implicated in hematopoiesis, angiogenesis, and leukemogenesis, contains IRES sequences in the 5' untranslated region. To clarify the roles of the IRES element in Runx1 function, we generated knock-in mice for either wild-type Runx1 or Runx1/Evi1, a Runx1 fusion protein identified in human leukemia. In both cases, native promoter-dependent transcription was retained, whereas IRES-mediated translation was eliminated. Interestingly, homozygotes expressing wild-type Runx1 deleted for the IRES element (Runx1(Delta IRES/Delta IRES)) died in utero with prominent dilatation of peripheral blood vessels due to impaired pericyte development. In addition, hematopoietic cells in the Runx1(Delta IRES/Delta IRES) fetal liver were significantly decreased, and exhibited an altered differentiation pattern, a reduced proliferative activity, and an impaired reconstitution ability. On the other hand, heterozygotes expressing Runx1/Evi1 deleted for the IRES element (Runx1(+/RE Delta IRES)) were born normally and did not show any hematological abnormalities, in contrast that conventional Runx1/Evi1 heterozygotes die in utero with central nervous system hemorrhage and Runx1/Evi1 chimeric mice develop acute leukemia. The findings reported here demonstrate the essential roles of the IRES element in Runx1 function under physiological and pathological conditions.


Subject(s)
5' Untranslated Regions , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoiesis/genetics , Leukemia/genetics , Neovascularization, Physiologic/genetics , Animals , Cell Differentiation , Gene Expression Regulation, Leukemic , Gene Knock-In Techniques , Heterozygote , Liver/pathology , Mice , Models, Animal , Ribosomes/metabolism
15.
Hepatology ; 52(3): 1089-99, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20623582

ABSTRACT

UNLABELLED: p130Cas, Crk-associated substrate (Cas), is an adaptor/scaffold protein that plays a central role in actin cytoskeletal reorganization. We previously showed that mice in which Cas was deleted (Cas(-/-)) died in utero because of early cardiovascular maldevelopment. To further investigate the in vivo roles of Cas, we generated mice with a hypomorphic Cas allele lacking the exon 2-derived region (Cas(Deltaex2/Deltaex2)), which encodes Src homology domain 3 (SH3) of Cas. Cas(Deltaex2/Deltaex2) mice again died as embryos, but they particularly showed progressive liver degeneration with hepatocyte apoptosis. Because Cas expression in the liver is preferentially detected in sinusoidal endothelial cells (SECs), the observed hepatocyte apoptosis was most likely ascribable to impaired function of SECs. To address this possibility, we stably introduced a Cas mutant lacking the SH3 domain (Cas DeltaSH3) into an SEC line (NP31). Intriguingly, the introduction of Cas DeltaSH3 induced a loss of fenestrae, the characteristic cell-penetrating pores in SECs that serve as a critical route for supplying oxygen and nutrients to hepatocytes. The disappearance of fenestrae in Cas DeltaSH3-expressing cells was associated with an attenuation of actin stress fiber formation, a marked reduction in tyrosine phosphorylation of Cas, and defective binding of Cas to CrkII. CONCLUSION: Cas plays pivotal roles in liver development through the reorganization of the actin cytoskeleton and formation of fenestrae in SECs.


Subject(s)
Cell Surface Extensions/physiology , Crk-Associated Substrate Protein/physiology , Endothelium/physiology , Liver/embryology , Liver/physiology , Actins/physiology , Animals , Apoptosis/physiology , Cell Line , Cell Surface Extensions/ultrastructure , Crk-Associated Substrate Protein/genetics , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Endothelium/cytology , Endothelium/ultrastructure , Exons/genetics , Female , Hepatocytes/cytology , Hepatocytes/physiology , Hepatocytes/ultrastructure , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Phosphorylation/physiology , Rats
16.
Genes Cells ; 14(12): 1383-94, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19930468

ABSTRACT

The activation mechanisms of Src family kinases (SFKs) involve the dissociation of the intramolecular interaction between the Src homology (SH) 3 and kinase domain. This process is mediated by the intermolecular attack of outer ligands to the SH3 domain. By using a yeast two-hybrid screen, we isolated a relevant ligand involved in the activation mechanisms of SFKs. This molecule was found to be identical to a recently recognized kinetochore protein--designated as centromere protein (CENP)-V--which is required for the progression of mitosis. We show here that human CENP-V plays further roles in cell dynamics; the proline-rich region of human CENP-V associates with the SH3 domains of SFKs and potently activates SFKs, whereas another domain of CENP-V that possesses a highly conserved cysteine array confers the ability to associate with stabilized microtubules (MTs). Human CENP-V distributes to the cell protrusion and to the leading edge of migrating cells in response to external stimuli, and depletion of CENP-V by RNA interference significantly attenuates closure of a scratch wound. These findings indicate that human CENP-V is involved in directional cell motility as well as in the progression of mitosis, as a scaffolding molecule that links MTs and SFKs.


Subject(s)
Centromere/metabolism , DNA-Binding Proteins/metabolism , Microtubules/metabolism , src Homology Domains/physiology , src-Family Kinases/metabolism , Amino Acid Sequence , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Enzyme Activation , Female , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Kinetochores , Molecular Sequence Data , RNA, Small Interfering/pharmacology , Sequence Homology, Amino Acid , Tissue Distribution , Tumor Cells, Cultured , Two-Hybrid System Techniques , Ubiquitination , Wound Healing
17.
Cancer Sci ; 100(7): 1219-26, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19432895

ABSTRACT

Chronic myelogenous leukemia (CML) is a hematological malignancy that begins as indolent chronic phase (CP) but inevitably progresses to fatal blast crisis (BC). p210BCR/ABL, a chimeric protein with enhanced kinase activity, initiates CML CP, and additional genetic alterations account for progression to BC, but the precise mechanisms underlying disease evolution are not fully understood. In the present study, we investigated the possible contribution of dysfunction of Bcl11b, a zinc-finger protein required for thymocyte differentiation, and of H2AX, a histone protein involved in DNA repair, to the transition from CML CP to BC. For this purpose, we crossed CML CP-exhibiting p210BCR/ABL transgenic (BA(tg/-)) mice with Bcl11b heterozygous (Bcl11b(+/-)) mice and H2AX heterozygous (H2AX(+/-)) mice. Interestingly, p210BCR/ABL transgenic, Bcl11b heterozygous (BA(tg/-)Bcl11b(+/-)) mice and p210BCR/ABL transgenic, H2AX heterozygous (BA(tg/-)H2AX(+/-)) mice frequently developed CML BC with T-cell phenotype and died in a short period. In addition, whereas p210BCR/ABL was expressed in all of the leukemic tissues, the expression of Bcl11b and H2AX was undetectable in several tumors, which was attributed to the loss of the residual normal allele or the lack of mRNA expression. These results indicate that Bcl11b and H2AX function as tumor suppressor and that haploinsufficiency and acquired loss of these gene products cooperate with p210BCR/ABL to develop CML BC.


Subject(s)
Blast Crisis/genetics , Histones/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Chromosome Aberrations , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Haplotypes/genetics , Heterozygote , Histones/metabolism , Humans , Mice , Mice, Transgenic , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism
18.
Blood ; 113(19): 4702-10, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19234145

ABSTRACT

Chronic myelogenous leukemia (CML) is a hematopoietic disorder originating from p210BCR/ABL-transformed stem cells, which begins as indolent chronic phase (CP) but progresses into fatal blast crisis (BC). To investigate molecular mechanism(s) underlying disease evolution, CML-exhibiting p210BCR/ABL transgenic mice were crossed with BXH2 mice that transmit a replication-competent retrovirus. Whereas nontransgenic mice in the BXH2 background exclusively developed acute myeloid leukemia, p210BCR/ABL transgenic littermates developed nonmyeloid leukemias, in which inverse polymerase chain reaction detected 2 common viral integration sites (CISs). Interestingly, one CIS was transgene's own promoter, which up-regulated p210BCR/ABL expression. The other was the 5' noncoding region of a transcription factor, Zfp423, which induced aberrant Zfp423 expression. The cooperative activities of Zfp423 and p210BCR/ABL were demonstrated as follows: (1) introduction of Zfp423 in p210BCR/ABL transgenic bone marrow (BM) cells increased colony-forming ability, (2) suppression of ZNF423 (human homologue of Zfp423) in ZNF423-expressing, p210BCR/ABL-positive hematopoietic cells retarded cell growth, (3) mice that received a transplant of BM cells transduced with Zfp423 and p210BCR/ABL developed acute leukemia, and (4) expression of ZNF423 was found in human BCR/ABL-positive cell lines and CML BC samples. These results demonstrate that enhanced expression of p210BCR/ABL and deregulated expression of Zfp423/ZNF423 contribute to CML BC.


Subject(s)
B-Lymphocytes/pathology , Blast Crisis/genetics , DNA-Binding Proteins/physiology , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Transcription Factors/physiology , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Colony-Forming Units Assay , DNA-Binding Proteins/antagonists & inhibitors , Female , Flow Cytometry , Gene Rearrangement , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Mice, Knockout , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Retroviridae , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transcription Factors/antagonists & inhibitors , Zinc Fingers
19.
Invest Ophthalmol Vis Sci ; 50(1): 242-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18791169

ABSTRACT

PURPOSE: In an earlier study, a cDNA was cloned that showed abundant expression in the eye at postnatal day (P)2 but was downregulated at P10; it was named ODAG (ocular development-associated gene). Its biological function was examined by generating and analyzing transgenic mice overexpressing ODAG (ODAG Tg) in the eye and by identifying ODAG-binding proteins. METHODS: Transgenic mice were generated by using the mouse Crx promoter. EGFP was designed to be coexpressed with transgenic ODAG, to identify transgene-expressing cells. Overexpression of ODAG was confirmed by Northern and Western blot analysis. IOP was measured with a microneedle technique. The eyes were macroscopically examined and histologically analyzed. EGFP expression was detected by confocal microscope. Proteins associated with ODAG were isolated by pull-down assay in conjugation with mass spectrometry. RESULTS: Macroscopically, ODAG Tg exhibited gradual protrusion of the eyeballs. The mean IOP of ODAG Tg was significantly higher than that of wild-type (WT) littermates. Histologic analysis exhibited optic nerve atrophy and impaired retinal development in the ODAG Tg eye. EGFP was expressed highly in the presumptive outer nuclear layer and weakly in the presumptive inner nuclear layer in the ODAG Tg retina. Rab6-GTPase-activating protein (Rab6-GAP) and its substrate, Rab6, were identified as ODAG-binding proteins. CONCLUSIONS: Deregulated expression of ODAG in the eye induces elevated intraocular pressure and optic nerve atrophy and impairs retinal development, possibly by interfering with the Rab6/Rab6-GAP-mediated signaling pathway. These results provide new insights into the mechanisms regulating ocular development, and ODAG Tg would be a novel animal model for human diseases caused by ocular hypertension.


Subject(s)
GATA1 Transcription Factor/genetics , Gene Expression Regulation/physiology , Intraocular Pressure , Ocular Hypertension/genetics , Optic Atrophy/genetics , Retinal Diseases/genetics , Animals , Blotting, Northern , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , GATA1 Transcription Factor/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mass Spectrometry , Mice , Mice, Transgenic , Ocular Hypertension/pathology , Optic Atrophy/pathology , Recombinant Fusion Proteins , Retinal Diseases/pathology , rab GTP-Binding Proteins
20.
Genes Cells ; 13(2): 145-57, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18233957

ABSTRACT

p130Cas (Cas, Crk-associated substrate) is an adaptor molecule composed of a Src homology 3 (SH3) domain, a substrate domain (SD) and a Src binding domain (SBD). The SH3 domain of Cas associates with focal adhesion kinase (FAK), but its role in cellular function has not fully been understood. To address this issue, we established and analyzed primary fibroblasts derived from mice expressing a truncated Cas lacking exon 2, which encodes the SH3 domain (Cas Deltaexon 2). In comparison to wild-type cells, Cas exon 2(Delta/Delta) cells showed reduced motility, which could be due to impaired tyrosine-phosphorylation of FAK and Cas, reduced FAK/Cas/Src/CrkII binding, and also impaired localization of Cas Deltaexon 2 to focal adhesions on fibronectin. In addition, to analyze downstream signaling pathways regulated by Cas exon 2, we performed microarray analyses. Interestingly, we found that a deficiency of Cas exon 2 up-regulated expression of CXC Chemokine Receptor-4 and CC Chemokine Receptor-5, which may be regulated by IkappaBalpha phosphorylation. These results indicate that the SH3-encoding exon of Cas participates in cell motility, tyrosine-phosphorylation of FAK and Cas, FAK/Cas/Src/CrkII complex formation, recruitment of Cas to focal adhesions and regulation of cell motility-associated gene expression in primary fibroblasts.


Subject(s)
Crk-Associated Substrate Protein/genetics , Animals , Cell Movement , Cells, Cultured , Crk-Associated Substrate Protein/chemistry , Crk-Associated Substrate Protein/metabolism , Exons , Fibroblasts/metabolism , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , Gene Expression , I-kappa B Proteins/metabolism , Mice , Mice, Knockout , Models, Biological , Multiprotein Complexes , NF-KappaB Inhibitor alpha , Proto-Oncogene Proteins c-crk/chemistry , Proto-Oncogene Proteins c-crk/metabolism , Receptors, CCR5/genetics , Receptors, CXCR4/genetics , Sequence Deletion , Signal Transduction , Thrombospondins/genetics , Wound Healing , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...