Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34663724

ABSTRACT

Although it is held that proinflammatory changes precede the onset of breast cancer, the underlying mechanisms remain obscure. Here, we demonstrate that FRS2ß, an adaptor protein expressed in a small subset of epithelial cells, triggers the proinflammatory changes that induce stroma in premalignant mammary tissues and is responsible for the disease onset. FRS2ß deficiency in mouse mammary tumor virus (MMTV)-ErbB2 mice markedly attenuated tumorigenesis. Importantly, tumor cells derived from MMTV-ErbB2 mice failed to generate tumors when grafted in the FRS2ß-deficient premalignant tissues. We found that colocalization of FRS2ß and the NEMO subunit of the IκB kinase complex in early endosomes led to activation of nuclear factor-κB (NF-κB), a master regulator of inflammation. Moreover, inhibition of the activities of the NF-κB-induced cytokines, CXC chemokine ligand 12 and insulin-like growth factor 1, abrogated tumorigenesis. Human breast cancer tissues that express higher levels of FRS2ß contain more stroma. The elucidation of the FRS2ß-NF-κB axis uncovers a molecular link between the proinflammatory changes and the disease onset.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Breast Neoplasms/immunology , Carcinogenesis , Cytokines/metabolism , Female , Humans , Inflammation/etiology , Inflammation/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , NF-kappa B/metabolism , Pregnancy , Receptor, ErbB-2/metabolism , Retroviridae Infections , Tumor Microenvironment/immunology , Tumor Virus Infections
2.
J Orthop Sci ; 27(6): 1315-1322, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34531086

ABSTRACT

BACKGROUND: Mutant isocitrate dehydrogenase (IDH) in chondrosarcoma produces the oncometabolite 2-hydroxyglutarate (2-HG) and contributes to malignant progression, and is therefore a potential therapeutic target for chondrosarcoma. Robust historical control data are important in clinical trials of rare cancers such as chondrosarcoma in order to show a clear benefit of new drugs. However, it remains controversial whether IDH mutation status is associated with the clinical outcome of chondrosarcoma, and this hinders the development of mutant IDH inhibitors in clinical trials.background METHODS: We investigated the relationship between IDH gene status and clinicopathological data in 38 chondrosarcoma patients from whom frozen tumor samples were obtained at the time of biopsy or surgery. Targeted next-generation sequencing was also performed to compare genetic alterations between patients with and without IDH mutations. METHODS RESULTS: The results revealed 15 cases (40%) of heterozygous IDH1 mutations and five cases (13%) of IDH2 mutations. IDH-mutant chondrosarcoma was associated with worse overall survival than IDH-wild-type chondrosarcoma (IDH1/2 Mut vs. IDH Wt, P = 0.006; IDH1 Mut vs. IDH Wt, P = 0.030; IDH2 Mut vs. IDH Wt, P < 0.0001). IDH mutation was also a significant poor prognostic factor both in univariate (P = 0.026) and multivariate (P = 0.048) analyses. Targeted next-generation sequencing revealed that characteristic mutations in chondrosarcoma, including TP53 and COL2A1, were more common in the IDH-mutant group than in the IDH-wild-type group.results CONCLUSION: This study is the first to report in detail the characteristics and clinical courses of IDH-mutant chondrosarcoma patients in Japan. Our data suggested that IDH-mutant chondrosarcomas might have a worse prognosis than that of IDH-wild-type chondrosarcoma, possibly through the more aggressive characters after metastasis. This information will be useful for designing clinical trials of mutant IDH inhibitors for treatment of advanced chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Humans , Isocitrate Dehydrogenase/genetics , Prognosis , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Mutation , Enzyme Inhibitors/pharmacology , Bone Neoplasms/genetics , Bone Neoplasms/pathology
3.
Cancer Sci ; 112(6): 2314-2324, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33792119

ABSTRACT

Mantle cell lymphoma (MCL) is a rare subtype of non-Hodgkin's lymphoma, which is characterized by overexpression of cyclin D1. Although novel drugs, such as ibrutinib, show promising clinical outcomes, relapsed MCL often acquires drug resistance. Therefore, alternative approaches for refractory and relapsed MCL are needed. Here, we examined whether a novel inhibitor of enhancer of zeste homologs 1 and 2 (EZH1/2), OR-S1 (a close analog of the clinical-stage compound valemetostat), had an antitumor effect on MCL cells. In an ibrutinib-resistant MCL patient-derived xenograft (PDX) mouse model, OR-S1 treatment by oral administration significantly inhibited MCL tumor growth, whereas ibrutinib did not. In vitro growth assays showed that compared with an established EZH2-specific inhibitor GSK126, OR-S1 had a marked antitumor effect on MCL cell lines. Furthermore, comprehensive gene expression analysis was performed using OR-S1-sensitive or insensitive MCL cell lines and showed that OR-S1 treatment modulated B-cell activation, differentiation, and cell cycle. In addition, we identified Cyclin Dependent Kinase Inhibitor 1C (CDKN1C, also known as p57, KIP2), which contributes to cell cycle arrest, as a direct target of EZH1/2 and showed that its expression influenced MCL cell proliferation. These results suggest that EZH1/2 may be a potential novel target for the treatment of aggressive ibrutinib-resistant MCL via CDKN1C-mediated cell cycle arrest.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Drug Resistance, Neoplasm/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma, Mantle-Cell/drug therapy , Piperidines/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Adenine/pharmacology , Adenine/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Mice , Piperidines/therapeutic use , Syndecan-1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancer Sci ; 111(12): 4371-4380, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32969571

ABSTRACT

Protein phosphatase 6 (PP6) is an essential serine/threonine protein phosphatase that acts as an important tumor suppressor. However, increased protein levels of PP6 have been observed in some cancer types, and they correlate with poor prognosis in glioblastoma. This raises a question about how PP6 protein levels are regulated in normal and transformed cells. In this study, we show that PP6 protein levels increase in response to pharmacologic and genetic inhibition of autophagy. PP6 associates with autophagic adaptor protein p62/SQSTM1 and is degraded in a p62-dependent manner. Accordingly, protein levels of PP6 and p62 fluctuate in concert under different physiological and pathophysiological conditions. Our data reveal that PP6 is regulated by p62-dependent autophagy and suggest that accumulation of PP6 protein in tumor tissues is caused at least partially by deficiency in autophagy.


Subject(s)
Autophagy/physiology , Phosphoprotein Phosphatases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Leupeptins/pharmacology , Macrolides/pharmacology , Male , Mice , Mice, Inbred C57BL , Proteolysis , RNA-Binding Proteins/metabolism , Sequestosome-1 Protein/metabolism
5.
Blood ; 131(16): 1833-1845, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29371181

ABSTRACT

Eradication of chemotherapy-resistant leukemia stem cells is expected to improve treatment outcomes in patients with acute myelogenous leukemia (AML). In a mouse model of AML expressing the MOZ-TIF2 fusion, we found that Ring1A and Ring1B, components of Polycomb repressive complex 1, play crucial roles in maintaining AML stem cells. Deletion of Ring1A and Ring1B (Ring1A/B) from MOZ-TIF2 AML cells diminished self-renewal capacity and induced the expression of numerous genes, including Glis2 Overexpression of Glis2 caused MOZ-TIF2 AML cells to differentiate into mature cells, whereas Glis2 knockdown in Ring1A/B-deficient MOZ-TIF2 cells inhibited differentiation. Thus, Ring1A/B regulate and maintain AML stem cells in part by repressing Glis2 expression, which promotes their differentiation. These findings provide new insights into the mechanism of AML stem cell homeostasis and reveal novel targets for cancer stem cell therapy.


Subject(s)
Gene Expression Regulation, Leukemic , Histone Acetyltransferases/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Nerve Tissue Proteins/biosynthesis , Nuclear Receptor Coactivator 2/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Polycomb Repressive Complex 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation , Histone Acetyltransferases/genetics , Kruppel-Like Transcription Factors/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Coactivator 2/genetics , Oncogene Proteins, Fusion/genetics , Polycomb Repressive Complex 1/genetics , Ubiquitin-Protein Ligases/genetics
6.
Cancer Sci ; 110(1): 194-208, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30343511

ABSTRACT

Multiple myeloma (MM) is an incurable hematological malignancy caused by accumulation of abnormal clonal plasma cells. Despite the recent development of novel therapies, relapse of MM eventually occurs as a result of a remaining population of drug-resistant myeloma stem cells. Side population (SP) cells show cancer stem cell-like characteristics in MM; thus, targeting these cells is a promising strategy to completely cure this malignancy. Herein, we showed that SP cells expressed higher levels of enhancer of zeste homolog (EZH) 1 and EZH2, which encode the catalytic subunits of Polycomb repressive complex 2 (PRC2), than non-SP cells, suggesting that EZH1 as well as EZH2 contributes to the stemness maintenance of the MM cells and that targeting both EZH1/2 is potentially a significant therapeutic approach for eradicating myeloma stem cells. A novel orally bioavailable EZH1/2 dual inhibitor, OR-S1, effectively eradicated SP cells and had a greater antitumor effect than a selective EZH2 inhibitor in vitro and in vivo, including a unique patient-derived xenograft model. Moreover, long-term continuous dosing of OR-S1 completely cured mice bearing orthotopic xenografts. Additionally, PRC2 directly regulated WNT signaling in MM, and overactivation of this signaling induced by dual inhibition of EZH1/2 eradicated myeloma stem cells and negatively affected tumorigenesis, suggesting that repression of WNT signaling by PRC2 plays an important role in stemness maintenance of MM cells. Our results show the role of EZH1/2 in the maintenance of myeloma stem cells and provide a preclinical rationale for therapeutic application of OR-S1, leading to significant advances in the treatment of MM.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Multiple Myeloma/prevention & control , Neoplastic Stem Cells/drug effects , Polycomb Repressive Complex 2/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Side-Population Cells/drug effects , Side-Population Cells/metabolism , Wnt Signaling Pathway/genetics
7.
Cancer Sci ; 109(8): 2342-2348, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29845708

ABSTRACT

Polycomb group (PcG) proteins regulate the expression of target genes by modulating histone modifications and are representative epigenetic regulators that maintain the stemness of embryonic and hematopoietic stem cells. Histone methyltransferases enhancer of zeste homolog 1 and 2 (EZH1/2), which are subunits of polycomb repressive complexes (PRC), are recurrently mutated or highly expressed in many hematological malignancies. EZH2 has a dual function in tumorigenesis as an oncogene and tumor suppressor gene, and targeting PRC2, in particular EZH1/2, for anticancer therapy has been extensively developed in the clinical setting. Here, we review the oncogenic function of EZH1/2 and introduce new therapeutic drugs targeting these enzymes.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Hematologic Neoplasms/genetics , Oncogenes/genetics , Polycomb Repressive Complex 2/genetics , Animals , Carcinogenesis/genetics , Humans , Polycomb-Group Proteins/genetics
8.
Immunity ; 31(4): 609-20, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19800266

ABSTRACT

Naturally arising regulatory T (Treg) cells express the transcription factor FoxP3, which critically controls the development and function of Treg cells. FoxP3 interacts with another transcription factor Runx1 (also known as AML1). Here, we showed that Treg cell-specific deficiency of Cbfbeta, a cofactor for all Runx proteins, or that of Runx1, but not Runx3, induced lymphoproliferation, autoimmune disease, and hyperproduction of IgE. Cbfb-deleted Treg cells exhibited impaired suppressive function in vitro and in vivo, with altered gene expression profiles including attenuated expression of FoxP3 and high expression of interleukin-4. The Runx complex bound to more than 3000 gene loci in Treg cells, including the Foxp3 regulatory regions and the Il4 silencer. In addition, knockdown of RUNX1 showed that RUNX1 is required for the optimal regulation of FoxP3 expression in human T cells. Taken together, our results indicate that the Runx1-Cbfbeta heterodimer is indispensable for in vivo Treg cell function, in particular, suppressive activity and optimal expression of FoxP3.


Subject(s)
Autoimmune Diseases/immunology , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/metabolism , Colon/immunology , Colon/pathology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/immunology , Forkhead Transcription Factors/immunology , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling , Immunoglobulin E/biosynthesis , Immunoglobulin E/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Stomach/immunology , Stomach/pathology , T-Lymphocytes, Regulatory/metabolism
9.
Stem Cells ; 34(5): 1151-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26822103

ABSTRACT

Cancer stem cells (CSCs) are believed to be maintained within a microenvironmental niche. Here we used polymer microarrays for the rapid and efficient identification of glioma CSC (GSC) niche mimicries and identified a urethane-based synthetic polymer, upon which two groups of niche components, namely extracellular matrices (ECMs) and iron are revealed. In cultures, side population (SP) cells, defined as GSCs in the rat C6 glioma cell line, are more efficiently sustained in the presence of their differentiated progenies expressing higher levels of ECMs and transferrin, while in xenografts, ECMs are supplied by the vascular endothelial cells (VECs), including SP cell-derived ones with distinctively greater ability to retain xenobiotics than host VECs. Iron is stored in tumor infiltrating host macrophages (Mφs), whose protumoral activity is potently enhanced by SP cell-secreted soluble factor(s). Finally, coexpression of ECM-, iron-, and Mφ-related genes is found to be predictive of glioma patients' outcome. Our polymer-based approach reveals the intrinsic capacities of GSCs, to adapt the environment to organize a self-advantageous microenvironment niche, for their maintenance and expansion, which redefines the current concept of anti-CSC niche therapy and has the potential to accelerate cancer therapy development. Stem Cells 2016;34:1151-1162.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Polymers/pharmacology , Stem Cell Niche , Tissue Scaffolds/chemistry , Animals , Brain Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Humans , Iron/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polyurethanes/pharmacology , Rats , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Side-Population Cells/cytology , Side-Population Cells/drug effects , Stem Cell Niche/drug effects , Stem Cell Niche/genetics , Transferrin/metabolism , Treatment Outcome
10.
Nucleic Acids Res ; 42(7): 4241-56, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24465000

ABSTRACT

Mixed-lineage leukemia (MLL) maintains the expression of cellular memory genes during development, while leukemic MLL fusion proteins aberrantly maintain expression of hematopoietic stem cell program genes such as HOXA9 to cause leukemia. However, the molecular mechanism of gene activation is unclear. Here we show that only two functional modules are necessary and sufficient for target recognition: those that bind to non-methylated CpGs and di-/tri-methylated histone H3 lysine 36 (H3K36me2/3). An artificial protein composed of the two targeting modules and an interaction domain for AF4-family coactivators can functionally substitute for MLL fusion proteins. Because H3K36me2/3 markers are indicative of active transcription, MLL fusion proteins target previously active CpG-rich genes and activate transcription by recruiting coactivators thereto. Our results indicate that such chromatin context-dependent gene activation is the fundamental mechanism by which MLL fusion proteins maintain the expression of the cellular memory/hematopoietic stem cell program genes.


Subject(s)
CpG Islands , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Histones/metabolism , Humans , Leukemia, Experimental/genetics , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/chemistry , Nucleosomes/metabolism , Oncogene Proteins, Fusion/chemistry , Protein Structure, Tertiary
11.
Proc Natl Acad Sci U S A ; 110(10): 3895-900, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431171

ABSTRACT

Monocytic leukemia zinc finger (MOZ)/KAT6A is a MOZ, Ybf2/Sas3, Sas2, Tip60 (MYST)-type histone acetyltransferase that functions as a coactivator for acute myeloid leukemia 1 protein (AML1)- and Ets family transcription factor PU.1-dependent transcription. We previously reported that MOZ directly interacts with p53 and is essential for p53-dependent selective regulation of p21 expression. We show here that MOZ is an acetyltransferase of p53 at K120 and K382 and colocalizes with p53 in promyelocytic leukemia (PML) nuclear bodies following cellular stress. The MOZ-PML-p53 interaction enhances MOZ-mediated acetylation of p53, and this ternary complex enhances p53-dependent p21 expression. Moreover, we identified an Akt/protein kinase B recognition sequence in the PML-binding domain of MOZ protein. Akt-mediated phosphorylation of MOZ at T369 has a negative effect on complex formation between PML and MOZ. As a result of PML-mediated suppression of Akt, the increased PML-MOZ interaction enhances p21 expression and induces p53-dependent premature senescence upon forced PML expression. Our research demonstrates that MOZ controls p53 acetylation and transcriptional activity via association with PML.


Subject(s)
Histone Acetyltransferases/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Base Sequence , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Knockout Techniques , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Humans , Intranuclear Inclusion Bodies/metabolism , Leukemia, Promyelocytic, Acute/genetics , Mice , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , p300-CBP Transcription Factors/metabolism
12.
Proc Natl Acad Sci U S A ; 110(11): 4339-44, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440206

ABSTRACT

Despite their nearly universal activation of mammalian target of rapamycin (mTOR) signaling, glioblastomas (GBMs) are strikingly resistant to mTOR-targeted therapy. We analyzed GBM cell lines, patient-derived tumor cell cultures, and clinical samples from patients in phase 1 clinical trials, and find that the promyelocytic leukemia (PML) gene mediates resistance to mTOR-targeted therapies. Direct mTOR inhibitors and EGF receptor (EGFR) inhibitors that block downstream mTOR signaling promote nuclear PML expression in GBMs, and genetic overexpression and knockdown approaches demonstrate that PML prevents mTOR and EGFR inhibitor-dependent cell death. Low doses of the PML inhibitor, arsenic trioxide, abrogate PML expression and reverse mTOR kinase inhibitor resistance in vivo, thus markedly inhibiting tumor growth and promoting tumor cell death in mice. These results identify a unique role for PML in mTOR and EGFR inhibitor resistance and provide a strong rationale for a combination therapeutic strategy to overcome it.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Nuclear Proteins/metabolism , Oxides/pharmacology , TOR Serine-Threonine Kinases/biosynthesis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Arsenic Trioxide , Cell Line, Tumor , ErbB Receptors/biosynthesis , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Male , Mice , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
13.
Cancer Sci ; 106(3): 227-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25529853

ABSTRACT

Acute myeloid leukemia is a clonal malignant disorder derived from a small number of leukemic stem cells (LSCs). Rearrangements of the mixed lineage leukemia (MLL) gene are found in acute myeloid leukemia associated with poor prognosis. The upregulation of Hox genes is critical for LSC induction and maintenance, but is unlikely to support malignancy and the high LSC frequency observed in MLL leukemias. The present study shows that MLL fusion proteins interact with the transcription factor PU.1 to activate the transcription of CSF-1R, which is critical for LSC activity. Acute myeloid leukemia is cured by either deletion of PU.1 or ablation of cells expressing CSF-1R. Kinase inhibitors specific for CSF-1R prolong survival time. These findings indicate that PU.1-mediated upregulation of CSF-1R is a critical effector of MLL leukemogenesis.


Subject(s)
Carcinogenesis/genetics , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Proto-Oncogene Proteins/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Trans-Activators/genetics , Animals , Gene Expression Regulation, Leukemic , Genes, Homeobox , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells , Phenylurea Compounds/pharmacology , Prognosis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Recombinant Fusion Proteins/genetics , Signal Transduction , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Thiazoles/pharmacology , Transcription, Genetic , Transcriptional Activation , Up-Regulation
14.
Rinsho Ketsueki ; 56(8): 1045-52, 2015 Aug.
Article in Japanese | MEDLINE | ID: mdl-26345565

ABSTRACT

Mutations in isocitrate dehydrogenase (IDH) 1 and 2 are frequently observed in acute myeloid leukemia (AML), glioma, and many other cancers. While wild-type IDHs mediate exchanges between isocitrate and α-ketoglutarate (α-KG), mutant IDHs convert α-KG to oncometabolite 2-hydroxyglutarate (2-HG), which causes dysregulation of a set of α-KG-dependent dioxygenases such as TET, histone demethylase and others. Because mutant IDH has no necessary functions in normal cells, inhibitors directed against mutant IDH are not expected to have the side effects as anti-cancer agents. To determine whether mutant IDH enzymes are valid targets for cancer therapy, we created a mouse model of mutant IDH2-dependent AML. By using a combination of AML model mice with cre-loxp, we conditionally deleted mutant IDH2 from AML mice, which resulted in the loss of leukemia stem cells and significantly delayed the progression of AML. These results indicate that mutant IDHs are promising targets for anticancer therapy.


Subject(s)
Hypoxia/genetics , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid/genetics , Mutation , Signal Transduction , Animals , Disease Models, Animal , Homeodomain Proteins/metabolism , Humans , Hypoxia/metabolism , Leukemia, Myeloid/metabolism , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism
15.
Cancer Sci ; 105(3): 315-23, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24397609

ABSTRACT

The CALM-AF10 fusion gene, which results from a t(10;11) translocation, is found in a variety of hematopoietic malignancies. Certain HOXA cluster genes and MEIS1 genes are upregulated in patients and mouse models that express CALM-AF10. Wild-type clathrin assembly lymphoid myeloid leukemia protein (CALM) primarily localizes in a diffuse pattern within the cytoplasm, whereas AF10 localizes in the nucleus; however, it is not clear where CALM-AF10 acts to induce leukemia. To investigate the influence of localization on leukemogenesis involving CALM-AF10, we determined the nuclear export signal (NES) within CALM that is necessary and sufficient for cytoplasmic localization of CALM-AF10. Mutations in the NES eliminated the capacity of CALM-AF10 to immortalize murine bone-marrow cells in vitro and to promote development of acute myeloid leukemia in mouse models. Furthermore, a fusion of AF10 with the minimal NES can immortalize bone-marrow cells and induce leukemia in mice. These results suggest that during leukemogenesis, CALM-AF10 plays its critical roles in the cytoplasm.


Subject(s)
Leukemia/metabolism , Monomeric Clathrin Assembly Proteins/physiology , Oncogene Proteins, Fusion/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , COS Cells , Carcinogenesis/genetics , Carcinogenesis/metabolism , Chlorocebus aethiops , Female , Leukemia/genetics , Leukemia/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Monomeric Clathrin Assembly Proteins/chemistry , Neoplasm Transplantation , Nuclear Export Signals , Tumor Cells, Cultured
16.
Cancer Sci ; 104(8): 1033-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23679839

ABSTRACT

AML1/RUNX1 is a frequent target of chromosome translocations and mutations in myeloid and B-cell leukemias, and upregulation of AML1 is also observed in some cases of T-cell leukemias and lymphomas. This study shows that the incidence of thymic lymphoma in p53-null mice is less frequent in the Aml1(+/-) than in the Aml1(+/+) background. AML1 is upregulated in p53-null mouse bone-marrow cells and embryonic fibroblasts. In the steady state, p53 binds to and inhibits the distal AML1 promoter. When the cells are exposed to stresses, p53 is released from the distal AML1 promoter, resulting in upregulation of AML1. Overexpression of AML1 stimulates T-lymphocyte proliferation. These results suggest that upregulation of AML1 induced by loss of p53 promotes lymphoid-cell proliferation, thereby inducing lymphoma development.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , T-Lymphocytes/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Growth Processes/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , T-Lymphocytes/metabolism , Thymus Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation
17.
J Cell Sci ; 124(Pt 13): 2208-19, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21670200

ABSTRACT

The mixed lineage leukemia (MLL) proto-oncogenic protein is a histone-lysine N-methyltransferase that is produced by proteolytic cleavage and self-association of the respective functionally distinct subunits (MLL(N) and MLL(C)) to form a holocomplex involved in epigenetic transcriptional regulation. On the basis of studies in Drosophila it has been suggested that the separated subunits might also have distinct functions. In this study, we used a genetically engineered mouse line that lacked MLL(C) to show that the MLL(N)-MLL(C) holocomplex is responsible for MLL functions in various developmental processes. The stability of MLL(N) is dependent on its intramolecular interaction with MLL(C), which is mediated through the first and fourth plant homeodomain (PHD) fingers (PHD1 and PHD4) and the phenylalanine/tyrosine-rich (FYRN) domain of MLL(N). Free MLL(N) is destroyed by a mechanism that targets the FYRN domain, whereas free MLL(C) is exported to the cytoplasm and degraded by the proteasome. PHD1 is encoded by an alternatively spliced exon that is occasionally deleted in T-cell leukemia, and its absence produces an MLL mutant protein that is deficient for holocomplex formation. Therefore, this should be a loss-of-function mutant allele, suggesting that the known tumor suppression role of MLL may also apply to the T-cell lineage. Our data demonstrate that the dissociated MLL subunits are subjected to distinct degradation pathways and thus not likely to have separate functions unless the degradation mechanisms are inhibited.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Leukemia, T-Cell/genetics , Mice , Mice, Knockout , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb-Group Proteins , Protein Processing, Post-Translational , Protein Subunits/genetics , Protein Subunits/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
18.
Gastroenterology ; 142(3): 572-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155184

ABSTRACT

BACKGROUND & AIMS: Loss of promyelocytic leukemia protein (PML) nuclear body (NB) formation has been reported in colorectal and other solid tumors. However, genetic alteration of PML is rarely observed in these tumors; the exact mechanisms that mediate loss of PML function are not known. METHODS: We previously used a comprehensive shotgun mass spectrometry approach to identify PML as 1 of 70 proteins that coimmunoprecipitate with anti-T-cell factor 4 in DLD-1 and HCT116 colorectal cancer cell lines; we investigated the effects of altered ß-catenin expression on PML function in these cells. RESULTS: ß-catenin specifically interacted with the product of PML transcript variant IV (PML-IV) through the armadillo repeat domain of ß-catenin. Overexpression of ß-catenin in colorectal cancer cells disrupted the subcellular compartmentalization of PML-IV, whereas knockdown of ß-catenin restored formation of PML-NB. Modification of PML by the small ubiquitin-related modifier (SUMO) is required for proper assembly of PML-NB. ß-catenin inhibited Ran-binding protein 2-mediated SUMOylation of PML-IV. CONCLUSIONS: ß-catenin interacts with PML isoform IV and disrupts PML-IV function and PML-NB formation by inhibiting Ran-binding protein 2-mediated SUMO modification of PML-IV. These findings indicate the involvement of a posttranslational mechanism in disruption of PML-NB organization in cancer cells and provide more information about the oncogenic functions of ß-catenin.


Subject(s)
Colorectal Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Lysine , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Binding , Protein Isoforms , RNA Interference , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Transcription Factor 4 , Transcription Factors/genetics , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
19.
Blood ; 118(9): 2443-53, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21753189

ABSTRACT

The histone acetyltransferases (HATs) of the MYST family include TIP60, HBO1, MOZ/MORF, and MOF and function in multisubunit protein complexes. Bromodomain-containing protein 1 (BRD1), also known as BRPF2, has been considered a subunit of the MOZ/MORF H3 HAT complex based on analogy with BRPF1 and BRPF3. However, its physiologic function remains obscure. Here we show that BRD1 forms a novel HAT complex with HBO1 and regulates erythropoiesis. Brd1-deficient embryos showed severe anemia because of impaired fetal liver erythropoiesis. Biochemical analyses revealed that BRD1 bridges HBO1 and its activator protein, ING4. Genome-wide mapping in erythroblasts demonstrated that BRD1 and HBO1 largely colocalize in the genome and target key developmental regulator genes. Of note, levels of global acetylation of histone H3 at lysine 14 (H3K14) were profoundly decreased in Brd1-deficient erythroblasts and depletion of Hbo1 similarly affected H3K14 acetylation. Impaired erythropoiesis in the absence of Brd1 accompanied reduced expression of key erythroid regulator genes, including Gata1, and was partially restored by forced expression of Gata1. Our findings suggest that the Hbo1-Brd1 complex is the major H3K14 HAT required for transcriptional activation of erythroid developmental regulator genes.


Subject(s)
Erythropoiesis , Histone Acetyltransferases/physiology , Liver/embryology , Protein Processing, Post-Translational , Trans-Activators/physiology , Acetylation , Anemia/embryology , Anemia/genetics , Animals , Carrier Proteins/physiology , DNA Damage , DNA Replication , Fetal Death/blood , Fetal Death/etiology , Fetal Death/genetics , GATA1 Transcription Factor/metabolism , Genes, Lethal , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , K562 Cells , Liver/physiology , Mice , Mice, Inbred C57BL , Multiprotein Complexes , Neoplasms/genetics , Neoplasms/metabolism , Protein Interaction Mapping , RNA, Small Interfering/pharmacology , Trans-Activators/deficiency , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/physiology
20.
Electrophoresis ; 34(11): 1670-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23483572

ABSTRACT

Nucleophosmin (NPM) is a novel prognostic biomarker for Ewing's sarcoma. To evaluate the prognostic utility of NPM, we conducted an interactomic approach to characterize the NPM protein complex in Ewing's sarcoma cells. A gene suppression assay revealed that NPM promoted cell proliferation and the invasive properties of Ewing's sarcoma cells. FLAG-tag-based affinity purification coupled with liquid chromatography-tandem mass spectrometry identified 106 proteins in the NPM protein complex. The functional classification suggested that the NPM complex participates in critical biological events, including ribosome biogenesis, regulation of transcription and translation, and protein folding, that are mediated by these proteins. In addition to JAK1, a candidate prognostic biomarker for Ewing's sarcoma, the NPM complex, includes 11 proteins known as prognostic biomarkers for other malignancies. Meta-analysis of gene expression profiles of 32 patients with Ewing's sarcoma revealed that 6 of 106 were significantly and independently associated with survival period. These observations suggest a functional role as well as prognostic value of these NPM complex proteins in Ewing's sarcoma. Further, our study suggests the potential applications of interactomics in conjunction with meta-analysis for biomarker discovery.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/metabolism , Nuclear Proteins , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Prognosis , Protein Binding , Protein Interaction Maps , RNA Interference , RNA, Small Interfering/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology
SELECTION OF CITATIONS
SEARCH DETAIL