Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Leuk Res ; 63: 1-9, 2017 12.
Article in English | MEDLINE | ID: mdl-29055789

ABSTRACT

Recombinant human soluble thrombomodulin (rTM) is a newly developed anti-coagulant approved for treatment of disseminated intravascular coagulation (DIC) in Japan. rTM exerts anti-inflammatory and cytoprotective functions via its lectin-like and epidermal growth factor-like domains, respectively. In this study, we retrospectively reviewed the treatment of 21 consecutive patients with coagulopathy, complicated by acute promyelocytic leukemia (APL), with all-trans retinoic acid (ATRA) with or without combination with rTM. Surprisingly, none of the 14 rTM-treated patients developed retinoic acid (RA)-related differentiation syndrome (DS). The co-culture of vascular endothelial cell-derived EA.hy926 and APL-derived NB4 cells in the presence of RA increased production of tumor necrosis factor alpha (TNF-α) in culture media, in parallel with activation of p38 mitogen-activated protein kinase and increased levels of intracellular adhesion molecule 1 (ICAM1) in EA.hy926 cells. This was also associated with increased levels of the phosphorylated forms of VE-cadherin and enhanced vascular permeability of EA.hy926 monolayers. Importantly, addition of rTM to this co-culture system inhibited the RA-induced phosphorylation of p38 and VE-cadherin and decreased ICAM1 and vascular permeability in EA.hy926 cells, without a decrease inthe levels of TNF-α. Taken together, use of rTM may be a promising treatment strategy to prevent DS in APL patients who receive ATRA.


Subject(s)
Cell Differentiation/drug effects , Disseminated Intravascular Coagulation/epidemiology , Leukemia, Promyelocytic, Acute/drug therapy , Respiratory Distress Syndrome/epidemiology , Thrombomodulin/metabolism , Tretinoin/adverse effects , Adult , Aged , Antineoplastic Agents/adverse effects , Apoptosis , Biomarkers, Tumor , Cell Proliferation , Coculture Techniques , Disseminated Intravascular Coagulation/chemically induced , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , In Vitro Techniques , Japan/epidemiology , Male , Middle Aged , Prognosis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respiratory Distress Syndrome/chemically induced , Retrospective Studies , Survival Rate , Thrombomodulin/genetics , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
2.
Leuk Res ; 58: 83-90, 2017 07.
Article in English | MEDLINE | ID: mdl-28501737

ABSTRACT

BCR/ABL tyrosine kinase inhibitors (TKIs) have significantly improved the prognosis for in dividuals with chronic myeloid leukemia (CML). However, many patients treated with TKIs suffer from TKI-related complications. In particular, vascular events such as peripheral artery occlusive disease have become aserious clinical problem for patients who receive the TKI, nilotinib. At present, the molecular mechanisms by which TKIs cause vascular endothelial cell insults remain unknown.This study explored the effects of the TKIs, imatinib, nilotinib and dasatinib, on vascular endothelial cells in vitro, and found that only nilotinib induced expression of interleukin-1ß (IL-1ß) by vascular endothelial cells. Nilotinib-induced IL-1ß expression stimulated the adhesion of monocytes to vascular endothelial cells in association with an increase in levels of adhesion molecules. MicroRNA database searching identified miR-3121-3p binding sites in the 3'-UTR of the IL-1ß gene. Exposure of endothelial cells to nilotinib caused downregulation of miR-3121-3p in these cells. Importantly, forced-expression of miR-3121-3p counteracted nilotinib-induced expression of IL-1ß. Importantly, serum levels if IL-1ß were significantly elevated in CML patients receiving nilotinib (n=14) compared to those receiving other TKIs (n=16) (3.76±1.22pg/ml vs 0.27±0.77pg/ml, p<0.05). Taken together, our data suggest that nilotinib decreases levels of miR-3121-3p resulting in an increase in expression of IL-1ß and adhesion molecules in vascular endothelial cells. The miR-3121-3p/IL-1ß axis could be a potential target to prevent vascular events in CML patients with high risk of vascular events.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Interleukin-1beta/biosynthesis , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Cell Line , Down-Regulation , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , MicroRNAs/biosynthesis , Real-Time Polymerase Chain Reaction
3.
Sci Rep ; 7(1): 692, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28386128

ABSTRACT

Thrombomodulin (TM) stimulates angiogenesis and protects vascular endothelial cells (ECs) via its fifth epidermal growth factor-like region (TME5); however, the cell surface receptor that mediates the pro-survival signaling activated by TM has remained unknown. We applied pull-down assay followed by MALDI-TOF MS and western blot analysis, and identified G-protein coupled receptor 15 (GPR15) as a binding partner of TME5. TME5 rescued growth inhibition and apoptosis caused by calcineurin inhibitor FK506 in vascular ECs isolated from wild type (WT) C57BL/6 mice. On the other hand, TME5 failed to protect ECs isolated from GPR15 knockout (GPR15 KO) mice from FK506-caused vascular injury. TME5 induced activation of extracellular signal-regulated kinase (ERK) and increased level of anti-apoptotic proteins in a GPR15 dependent manner. In addition, in vivo Matrigel plug angiogenesis assay found that TME5 stimulated angiogenesis in mice. TME5 promoted endothelial migration in vitro. Furthermore, TME5 increased production of NO in association with activated endothelial NO synthase (eNOS) in ECs. All these pro-angiogenesis functions of TME5 were abolished by knockout of GPR15. Our findings suggest that GPR15 plays an important role in mediating cytoprotective function as well as angiogenesis of TM.


Subject(s)
Cytoprotection/genetics , Neovascularization, Physiologic/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Thrombomodulin/metabolism , Animals , Cytokines/metabolism , Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Molecular Weight , Nitric Oxide/metabolism , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
4.
Biol Blood Marrow Transplant ; 23(5): 746-756, 2017 May.
Article in English | MEDLINE | ID: mdl-28167153

ABSTRACT

Thrombomodulin (TM) exerts anti-inflammatory functions. We previously found that recombinant human soluble TM alleviated murine graft-versus-host disease (GVHD). Nevertheless, it is unclear how TM mediates its anti-inflammatory functions in GVHD. Here, we identified G-protein coupled receptor 15 (GPR15) expressed on T cells as a receptor/sensor of TM. The fifth region of epidermal growth factor-like domain of TM (TME5) bound GPR15 in vitro. TME5 prolonged survival of mice undergoing acute GVHD after allogeneic hematopoietic stem cell transplantation (allo-HSCT). TME5 increased regulatory T cells (Tregs) but decreased Th 1 proportions in targeted organs. TME5 suppressed allo-reaction in vitro in association with an increase in the number of induced Tregs. However, the anti-inflammatory function of TME5 was abolished when GPR15 knockout T cells were used as donor T cells. We further found that TME5 suppressed production of IL-6 in T cells, which probably facilitated differentiation of Tregs. Moreover, TME5 reduced activation of bone marrow-derived dendritic cells (BMDCs) and hampered function of BMDCs in inducing allo-reaction in vivo and in vitro. Our findings suggested that inducing Tregs as well as blocking activation of DCs in vivo by using TME5 is a potential therapeutic option for preventing GVHD in allo-HSCT.


Subject(s)
Graft vs Host Disease/prevention & control , Peptide Fragments/pharmacology , Receptors, G-Protein-Coupled/physiology , Thrombomodulin/therapeutic use , Animals , Dendritic Cells/drug effects , Epidermal Growth Factor , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Inflammation/drug therapy , Leukocyte Reduction Procedures/methods , Mice , Peptide Fragments/therapeutic use , T-Lymphocytes, Regulatory/cytology , Transplantation, Homologous
5.
Cancer Sci ; 108(4): 685-695, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28107581

ABSTRACT

We previously showed that the CD82/signal transducer and activator of transcription/interleukin-10 (IL-10) axis is activated in CD34+ /CD38- AML cells that favor the bone marrow microenvironment. The present study explored the novel biological function of IL-10 in regulation of expression of adhesion molecules in AML cells and found that exposing AML cells to IL-10 induced expression of E-cadherin, but not other adhesion molecules, including VLA4, CD29, and LFA1. Downregulation of E-cadherin with an siRNA suppressed the adhesion of leukemia cells to bone marrow-derived mesenchymal stem cells and enhanced the anti-leukemia effect of cytarabine. A microRNA (miRNA) database search identified an miR-9 as a candidate miRNA binding onto the 3'-UTR of E-cadherin and regulating its expression. Notably, treatment of leukemia cells with IL-10 decreased miR-9 expression through hypermethylation of the miR-9 CpG islands. In addition, downregulation of DNA methyltransferase 3A by siRNAs decreased E-cadherin expression in parallel with an increase in levels of miR-9 in leukemia cells. Notably, short hairpin RNA-mediated IL-10 downregulation impaired engraftment of human AML cells and enhanced the anti-leukemia effect of cytarabine in conjunction with miR-9 upregulation and E-cadherin downregulation in a human AML xenograft model. Taken together, the IL-10/E-cadherin axis may be a promising therapeutic target for treating AML.


Subject(s)
Cadherins/genetics , Gene Expression Regulation, Leukemic/drug effects , Interleukin-10/pharmacology , Leukemia, Myeloid/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Acute Disease , Animals , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cells, Cultured , CpG Islands/genetics , Cytarabine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methyltransferase 3A , Humans , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred NOD , Mice, Knockout , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Xenograft Model Antitumor Assays
6.
Thromb Haemost ; 117(3): 570-579, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28078348

ABSTRACT

Thrombomodulin (TM) exerts cytoprotection via the fifth region of epidermal growth factor (EGF)-like domain of TM (TME5) by interacting with G-protein coupled receptor 15 (GPR15) expressed on cell surface of vascular endothelial cells. TM is also implied to mediate anti-inflammatory functions by unknown mechanism. By applying a lipopolysaccharide (LPS)-induced murine sepsis model, we assessed the role of TME5 in septic inflammation and coagulation. We found that TME5 treatment protected mice in association with ameliorating inflammation and coagulopathy in LPS-induced sepsis. Further study confirmed that TME5 bound GPR15 in vitro. Knock out of GPR15 abolished protective role of TME5 in sepsis model. GPR15 mediated anti-inflammatory function of TME5 through suppression of phosphorylation of IκBα, nuclear translocation of NF-κB and release of pro-inflammatory cytokines in macrophages (Macs). Knock out of GPR15 resulted in dysregulated immune response of Macs, characterised by excessive expression of pro-inflammatory genes and failing to limit immune response. This study indicates that TME5 exerts anti-inflammatory function through inhibition of NF-κB in a GPR15-dependent manner. The use of TME5 may be a potential therapeutic option for treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides , Macrophages/drug effects , Peptide Fragments/pharmacology , Receptors, G-Protein-Coupled/metabolism , Sepsis/prevention & control , Thrombomodulin , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Blood Coagulation/drug effects , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Domains , RAW 264.7 Cells , Receptors, G-Protein-Coupled/genetics , Sepsis/chemically induced , Sepsis/metabolism , Sepsis/pathology , Signal Transduction/drug effects , Thrombomodulin/chemistry , Thrombomodulin/metabolism , Time Factors
7.
Leuk Res ; 45: 24-32, 2016 06.
Article in English | MEDLINE | ID: mdl-27070757

ABSTRACT

In this study, we showed that the levels of EZH2 in bone marrow mononuclear cells (BMMNCs) isolated from individuals with chronic myeloid leukemia (CML) (n=12) were significantly greater than those in BMMNCs isolated from healthy volunteers (n=6) as well as individuals with Philadelphia chromosome-negative myeloproliferative neoplasms. Lentiviral transduction of the BCR/ABL gene in Ba/F3 cells increased EZH2 levels in parallel with phosphorylation of STAT5. Notably, chromatin immunoprecipitation assays showed that STAT5A bound to a promoter region of the EZH2 gene, resulting in an increase in the transcriptional activity of EZH2 in leukemia cells. Importantly, downregulation of EZH2 by short hairpin RNAs (shRNAs) inhibited the expression of XIAP and increased the miR-219 levels associated with a decrease in hypermethylation of miR-219-1 CpG islands. Moreover, overexpression of miR-219 decreased the levels of XIAP in CML cells. Since the 3'-untranslated region (3'-UTR) of XIAP contains miR219-5p-complementary binding site, miR-219 might modulate the expression of XIAP through binding of miR-219 on the 3'-UTR of XIAP. Taken together, BCR/ABL positively regulates the expression of EZH2 via STAT5 signaling. EZH2 modulates epigenetic changes at DNA methylated regions encoding miR-219 and downregulates the level of miR-219, resulting in upregulation of XIAP.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Fusion Proteins, bcr-abl/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mononuclear Phagocyte System/pathology , Bone Marrow Cells/pathology , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Gene Expression Regulation, Leukemic/drug effects , Humans , MicroRNAs/genetics , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , X-Linked Inhibitor of Apoptosis Protein/biosynthesis
8.
Cancer Med ; 4(9): 1426-31, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26139471

ABSTRACT

We recently found that CD82 inhibits matrix metalloproteinase 9 and augments adhesion of CD34(+) /CD38(-) acute myelogenous leukemia (AML) cells to the bone marrow (BM) microenvironment. The present study found that the use of an anti-CD82 monoclonal antibody (CD82 mAb) mobilized CD34(+) leukemia cells from BM into the peripheral blood in a humanized AML murine model. The use of CD82 mAb in combination with cytarabine (AraC) significantly prolonged survival of immunodeficient mice-bearing human AML cells than did treatment with either AraC or CD82 mAb alone. Taken together, the combination of an anti-leukemic agent and the mobilizing agent CD82 mAb may be a promising treatment strategy to treat patients with AML.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Kangai-1 Protein/antagonists & inhibitors , Leukemia/metabolism , Leukemia/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, CD34 , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Cytarabine/administration & dosage , Disease Models, Animal , Drug Synergism , Humans , Leukemia/drug therapy , Leukemia/mortality , Mice , Mice, Transgenic , Xenograft Model Antitumor Assays
9.
Leuk Res ; 39(8): 897-905, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26059451

ABSTRACT

Signal transducer and activator of transcription 5 (STAT5) is activated in CD34(+)/CD38(-) acute myelogenous leukemia (AML) cells. Inhibition of STAT5 induced apoptosis and sensitized these cells to the growth inhibition mediated by conventional chemotherapeutic agents. The present study attempted to identify molecules that are regulated by STAT5 in CD34(+)/CD38(-) AML cells by utilizing cDNA microarrays, comparing the gene expression profiles of control and STAT5A shRNA-transduced CD34(+)/CD38(-) AML cells. Interestingly, DNA methyltransferase (DNMT) 3A was downregulated after depletion of STAT5A in CD34(+)/CD38(-) AML cells. Reporter gene assays found that an increase in activity of DNMT3A occurred in response to activation of STAT5A in leukemia cells. On the other hand, dephosphorylation of STAT5A by AZ960 decreased this transcriptional activity. Further studies utilizing a chromatin immunoprecipitation assay identified a STAT5A-binding site on the promoter region of DNMT3A gene. Forced expression of STAT5A in leukemia cells caused hypermethylation on the promoter region of the tumor suppressor gene, PTEN, and downregulated its mRNA levels, as measured by methylation-specific and real-time polymerase chain reaction, respectively. Taken together, these data suggest that STAT5A positively regulates levels of DNMT3A, resulting in inactivation of tumor suppressor genes by epigenetic mechanisms in AML cells.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Leukemia, Myeloid, Acute/genetics , STAT5 Transcription Factor/physiology , Tumor Suppressor Proteins/physiology , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , DNA Methyltransferase 3A , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Leukemic/drug effects , Genes, Tumor Suppressor/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , RNA, Small Interfering/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Suppressor Proteins/antagonists & inhibitors
10.
PLoS One ; 10(5): e0125017, 2015.
Article in English | MEDLINE | ID: mdl-25955299

ABSTRACT

PURPOSE: We recently found that the tetraspanin family member, CD82, which is aberrantly expressed in chemotherapy-resistant CD34(+)/CD38- acute myelogenous leukemia (AML) cells, negatively regulates matrix metalloproteinase 9, and plays an important role in enabling CD34(+)/CD38(-) AML cells to adhere to the bone marrow microenvironment. This study explored novel functions of CD82 that contribute to AML progression. MATERIALS AND METHODS: We employed microarray analysis comparing the gene expression profiles between CD34(+)/CD38(-) AML cells transduced with CD82 shRNA and CD34(+)/CD38(-) AML cells transduced with control shRNA. Real-time RT-PCR and western blot analysis were performed to examine the effect of CD82 knockdown on the expression of the polycomb group member, enhancer of zeste homolog 2 (EZH2), in leukemia cells. A chromatin immunoprecipitation assay was performed to examine the effect of CD82 expression on the amount of EZH2 bound to the promoter regions of tumor suppressor genes in leukemia cells. We also utilized methylation-specific PCR to examine whether CD82 expression influences the methylation status of the tumor suppressor gene promoter regions in leukemia cells. RESULTS: Microarray analysis revealed that levels of EZH2 decreased after shRNA-mediated depletion of CD82 in CD34(+)/CD38(-) AML cells. Moreover, the antibody-mediated blockade of CD82 in leukemia cells lowered EZH2 expression via activation of p38 MAPK signaling, decreased the amount of EZH2 bound to the promoter regions of the tumor suppressor genes, and inhibited histone H3 lysine 27 trimethylation in these promoter regions, resulting in upregulation of the tumor suppressors at both the mRNA and protein levels.


Subject(s)
Kangai-1 Protein/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System , Polycomb Repressive Complex 2/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Cell Line, Tumor , DNA Methylation , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , Enzyme Activation , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Histones/metabolism , Humans , Kangai-1 Protein/genetics , Lysine/metabolism , Male , Models, Biological , PTEN Phosphohydrolase/genetics , Polycomb Repressive Complex 2/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism
11.
Cancer Sci ; 105(3): 297-307, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24350829

ABSTRACT

This study found that long-term exposure of chronic myelogenous leukemia (CML) K562 cells to BCR/ABL thyrosine kinase inhibitors (TKI) caused drug-resistance in association with an increase in levels of DNA methyltransferases (DNMT) and a decrease in levels of microRNA miR-217. These observations are clinically relevant; an increase in levels of DNMT3A in association with downregulation of miR-217 were noted in leukemia cells isolated from individuals with BCR/ABL TKI-resistant Philadelphia chromosome positive acute lymphoblastic leukemia (Ph(+) ALL) and CML. Further studies with TKI-resistant K562 cells found that forced expression of miR-217 inhibited expression of DNMT3A through a miR-217-binding site within the 3'-untranslated region of DNMT3A and sensitized these cells to growth inhibition mediated by the TKI. Of note, long-term exposure of K562 cells to dasatinib (10 nM) together with 5-Aza-2'-deoxycytidine (5-AzadC) (0.1 µM) potently inhibited proliferation of these cells in association with upregulation of miR-217 and downregulation of DNMT3A in vitro. In addition, a decrease in levels of DNMT3A and an increase in levels of miR-217 were noted in K562 tumors growing in immune-deficient mice that were treated with the combination of 5-AzadC and dasatinib. Taken together, Ph(+) leukemia cells acquire TKI resistance via downregulation of miR-217 and upregulation of DNMT3A. Inhibition of DNMT3A by forced expression of miR-217 or 5-AzadC may be useful to prevent drug resistance in individuals who receive TKI.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , MicroRNAs/genetics , Philadelphia Chromosome , Protein Kinase Inhibitors/pharmacology , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Survival/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Dasatinib , Decitabine , Down-Regulation , Drug Resistance, Neoplasm , Female , Gene Expression , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Pyrimidines/pharmacology , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 134(1): 55-64, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23797738

ABSTRACT

We recently reported that adhesion molecule CD82 is aberrantly expressed in CD34(+) /CD38(-) leukemia stem cells (LSCs). Here, we report the results of a functional analysis of CD82 in CD34(+) /CD38(-) acute myelogenous leukemia (AML) cells. Short hairpin (sh)RNA-mediated downregulation of CD82 resulted in a decrease in the level of IL-10. In contrast, forced expression of CD82 in CD34(+)/CD38(+) AML cells by transduction with CD82-expressing lentiviral particles resulted in an increase in the levels of IL-10. Notably, exposure of CD34(+)/CD38(-) AML cells to IL-10 stimulated clonogenic growth of these cells. Moreover, downregulation of CD82 by a shRNA dephosphorylated STAT5 in CD34(+)/CD38(-) AML cells. On the other hand, forced expression of CD82 resulted in increase in the levels of p-STAT5 in CD34(+)/CD38(+) AML cells. Chromatin immunoprecipitation (ChIP) assay results indicated that STAT5A binds to the promoter region of the IL-10 gene, while reporter gene assay results indicated stimulation of IL-10 expression at the transcriptional level. These results suggest that CD82 positively regulates the STAT5/IL-10 signaling pathway. Moreover, shRNA-mediated downregulation of CD82 expression in CD34(+)/CD38(-) AML cells dephosphorylated STAT5 in immunodeficient mice. Taken together, our data suggest that the CD82/STAT5/IL-10 signaling pathway is involved in the survival of CD34(+)/CD38(-) AML cells and may thus be a promising therapeutic target for eradication of AML LSCs.


Subject(s)
Interleukin-10/metabolism , Kangai-1 Protein/metabolism , Leukemia, Myeloid, Acute/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cell Line, Tumor , Cell Separation , Cell Survival , Chromatin Immunoprecipitation , Flow Cytometry , Heterografts , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Int J Cancer ; 134(5): 1183-94, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-23934627

ABSTRACT

This study explored molecular mechanisms by which Bcr-Abl induced expression of Aurora kinase A and B (AURKA and AURKB) in chronic myeloid leukemia cells. Lentiviral transduction of Bcr-Abl into either Ba/F3 or CD34(+) hematopoietic stem/progenitor cells potently increased levels of AURKA and AURKB in association with phosphorylation of AKT and stimulated their proliferation. Bcr-Abl-mediated expression of AURKA and AURKB were decreased in CD34(+) HSPCs when AKT was inactivated by an shRNA against AKT, suggesting that Bcr-Abl induced expression of AURKA and AURKB via AKT signaling. MLN8237, an inhibitor of AURKA, significantly inhibited the proliferation of freshly isolated CD34(+) CML cells in a dose-dependent manner as measured by colony forming assay. Importantly, inhibition of AURKA in CD34(+) leukemia cells freshly isolated from individuals with blast crisis of CML with Bcr-Abl T315I mutant (n = 2) by MLN8237 significantly impaired the engraftment of these cells in severely immunocompromised mice and decreased the weight of spleens. Taken together, Bcr-Abl induces expression of AURKA and AURKB at least in part via AKT. Inhibition of AURKA could be useful to overcome imatinib-resistance mediated by Bcr-Abl mutants.


Subject(s)
Aurora Kinase A/metabolism , Aurora Kinase B/metabolism , Fusion Proteins, bcr-abl/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , Animals , Azepines/pharmacology , Cell Proliferation/drug effects , Enzyme Activation , Fusion Proteins, bcr-abl/genetics , Humans , Lentivirus/genetics , Mice , Pyrimidines/pharmacology
14.
Apoptosis ; 18(11): 1403-1415, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23760752

ABSTRACT

Exposure of acute promyelocytic leukemia (APL) cells to all-trans retinoic acid (ATRA) increases levels of Mcl-1, however, the implication of ATRA-mediated expressions of Mcl-1 in these cells remains to be fully elucidated. This study found that exposure of NB4 and PL-21 cells to ATRA increased levels of Mcl-1 in association with phosphorylation of c-jun N-terminus kinases. Down-regulation of Mcl-1 by a small interfering (siRNA) or an inhibitor of JNK significantly potentiated the ability of ATRA to induce differentiation and apoptosis in these cells. On the other hand, the anti-leukemia effects of ATRA were blunted when Mcl-1 was forced expressed in NB4 and PL-21 cells as well as leukemia cells isolated from individuals with APL. Furthermore, down-regulation of Mcl-1 by an siRNA sensitized non-APL U937 and KG-1 leukemia cells to ATRA-mediated differentiation and apoptosis. Taken together, inhibition of Mcl-1 might be useful to potentiate the action of ATRA in APL as well as non-APL AML cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Leukemic , Leukemia, Promyelocytic, Acute/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Tretinoin/pharmacology , Cell Differentiation , Cell Proliferation , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
15.
Int J Cancer ; 133(11): 2706-19, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-23686525

ABSTRACT

We previously showed that Aurora kinase A (AURKA) is aberrantly expressed in acute myelogenous leukemia (AML) cells when compared to bone marrow mononuclear cells isolated from healthy volunteers. We have also shown that CD34(+) /CD38(-) AML cells, one of compartments enriched for leukemia stem cells in most leukemia subgroups, were relatively resistant to cytarabine-mediated growth inhibition when compared to their CD34(+) /CD38(+) counterparts. Our study attempted to identify therapeutic targets in CD34(+) /CD38(-) AML cells and found that CD34(+) /CD38(-) AML cells isolated from patients (n = 26) expressed larger amounts of AURKA than their CD34(+) /CD38(+) counterparts and CD34(+) normal hematopoietic stem/progenitor cells isolated from healthy volunteers (n = 6), as measured by real-time reverse-transcriptase polymerase chain reaction. Blockade of AURKA by the specific inhibitor MLN8237 or a short hairpin RNA (shRNA) against AURKA significantly inhibited proliferation, impaired self-renewal capability and induced apoptosis of CD34(+) /CD38(-) AML cells, in association with modulation of levels of Bcl-2 family member proteins. Importantly, inhibition of AURKA in CD34(+) /CD38(-) AML cells by MLN8237 or an shRNA significantly impaired engraftment of these cells in severely immunocompromised mice and appeared to prolong their survival. These results suggest that AURKA is a promising molecular target to eliminate chemotherapy-resistant CD34(+) /CD38(-) AML cells.


Subject(s)
Aurora Kinase A/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , ADP-ribosyl Cyclase 1/genetics , Animals , Antigens, CD34/genetics , Apoptosis/drug effects , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/biosynthesis , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Molecular Targeted Therapy , Pyrimidines/pharmacology
16.
Int J Cancer ; 133(8): 1967-81, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23564444

ABSTRACT

We previously showed that CD34⁺/CD38⁻ acute myelogenous leukemia (AML) cells, which contain leukemia stem cells, expressed a greater amount of the phosphorylated forms of JAK2 and STAT5 (p-JAK2 and p-STAT5) than their CD34⁺/CD38⁺ counterparts. To identify candidate cytokines that are involved in the activation of JAK2/STAT5 in CD34⁺/CD38⁻ AML cells, we compared the cytokine expression profiles of CD34⁺/CD38⁻ AML cells and their CD34⁺/CD38⁺ counterparts. Interestingly, freshly isolated CD34⁺/CD38⁻ AML cells from patients (n = 17) expressed less interleukin-1ß (IL-1ß) than their CD34⁺/CD38⁺ counterparts and CD34⁺ normal hematopoietic stem/progenitor cells from healthy volunteers (n = 6), as measured by real-time Reverse Transcription-Polymerase Chain Reaction (RT-PCR). Methylation-specific PCR found that IL-1B gene expression was silenced by methylation of the promoter region. Importantly, exposure of CD34⁺/CD38⁻ AML cells to IL-1ß (100 ng/ml) stimulated cell-cycle progression, induced apoptosis and sensitized these cells to growth inhibition by antileukemia agents. These changes occurred in conjunction with the downregulation of cyclin-dependent kinase inhibitor p21waf1, antiapoptotic proteins and p-STAT5. Forced expression of IL-1ß in CD34⁺/CD38⁻ AML cells by lentiviral transduction significantly impaired the self-renewal capacity of the cells and induced apoptosis. Additionally, when these CD34⁺/CD38⁻ AML cells with forced expression of IL-1ß were transplanted into severely immunocompromised mice, the engraftment of the cells and reconstitution of AML were significantly impaired. Taken together, our results indicate that the inhibition of STAT5 by IL-1ß may be a promising treatment strategy to eradicate leukemia stem cells in AML.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Leukemia, Myeloid, Acute/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , DNA Methylation , Down-Regulation , Humans , Janus Kinase 2/metabolism , Leukemia, Myeloid, Acute/drug therapy , Mice , Mice, Inbred NOD , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Promoter Regions, Genetic/genetics , STAT5 Transcription Factor/metabolism , Tumor Cells, Cultured
17.
J Org Chem ; 78(6): 2763-8, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23438033

ABSTRACT

The first synthesis of hexa(furan-2-yl)benzene derivatives is described. The RhCl3/i-Pr2NEt-catalyzed cyclotrimerization of di(furan-2-yl)acetylenes was an effective method for constructing hexa(furan-2-yl)benzene derivatives in good yields. Their π-extended derivatives were also synthesized by Suzuki-Miyaura coupling between hexakis(5-Bpinfuran-2-yl)benzene (Bpin = (pinacolato)boryl) and several aryl iodides.

18.
Immunology ; 138(3): 216-27, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23278479

ABSTRACT

Adult T-cell leukaemia-lymphoma (ATLL) is an aggressive malignancy of CD4(+)  CD25(+) T lymphocytes, characterized by a severely compromised immunosystem, in which the human T-cell lymphotropic virus type 1 (HTLV-1) has been recognized as the aetiological agent. This study found that an IκB kinase ß (IKKß) inhibitor Bay11-7082 inactivated mammalian target of rapamycin (mTOR), signal transducer and activator of transcription 3 and transcription factor nuclear factor-κB in HTLV-1-infected T cells; this was significantly enhanced in the presence of the mTOR inhibitor everolimus. In addition, Bay11-7082 decreased production of the immunosuppressive cytokine interleukin-10 (IL-10), which was further down-regulated when Bay11-7082 was combined with evelolimus in HTLV-1-infected T and ATLL cells isolated from patients. Interleukin-10 is known to inhibit maturation and the antigen-presenting function of dendritic cells (DCs). The culture media of HTLV-1-infected MT-1 cells, which contained a large amout of IL-10, hampered tumour necrosis factor-α-induced maturation of DCs isolated from healthy volunteers. Culture supernatant of MT-1 cells treated with a combination of Bay11-7082 and everolimus augmented maturation of DCs in association with a decrease in production of IL-10 and enhanced the allostimulatory function of DCs. Similarly, when DCs isolated from patients with ATLL were treated with the combination of Bay11-7082 and everolimus, they were fully matured and their capability to stimulate proliferation of lymphocytes was augmented. Taken together, the combination of Bay11-7082 and everolimus might exhibit immunostimulatory properties in HTLV-1-infected T and ATLL cells isolated from patients, and this combination may be potentially therapeutic to regain the compromised immunosystem in ATLL patients.


Subject(s)
Gene Expression Regulation/drug effects , HTLV-I Infections/immunology , I-kappa B Kinase/antagonists & inhibitors , Interleukin-10/genetics , Sirolimus/analogs & derivatives , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antineoplastic Agents/pharmacology , Cell Line , Dendritic Cells/cytology , Dendritic Cells/drug effects , Everolimus , HTLV-I Infections/genetics , Humans , I-kappa B Kinase/metabolism , Interleukin-10/metabolism , Lymphocyte Culture Test, Mixed , Monocytes/cytology , Monocytes/drug effects , Nitriles/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Sulfones/pharmacology , T-Lymphocytes/virology , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Int J Cancer ; 132(9): 2006-19, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23055153

ABSTRACT

To identify molecular targets in leukemia stem cells (LSCs), this study compared the protein expression profile of freshly isolated CD34(+) /CD38(-) cells with that of CD34(+) /CD38(+) counterparts from individuals with acute myelogenous leukemia (n = 2, AML) using isobaric tags for relative and absolute quantitation (iTRAQ). A total of 98 proteins were overexpressed, while six proteins were underexpressed in CD34(+) /CD38(-) AML cells compared with their CD34(+) /CD38(+) counterparts. Proteins overexpressed in CD34(+) /CD38(-) AML cells included a number of proteins involved in DNA repair, cell cycle arrest, gland differentiation, antiapoptosis, adhesion, and drug resistance. Aberrant expression of CD82, a family of adhesion molecules, in CD34(+) /CD38(-) AML cells was noted in additional clinical samples (n = 12) by flow cytometry. Importantly, down-regulation of CD82 in CD34(+) /CD38(-) AML cells by a short hairpin RNA (shRNA) inhibited adhesion to fibronectin via up-regulation of matrix metalloproteinases 9 (MMP9) and colony forming ability of these cells as assessed by transwell assay, real-time RT-PCR, and colony forming assay, respectively. Moreover, we found that down-regulation of CD82 in CD34(+) /CD38(-) AML cells by an shRNA significantly impaired engraftment of these cells in severely immunocompromised mice. Taken together, aberrant expression of CD82 might play a role in adhesion of LSCs to bone marrow microenvironment and survival of LSCs. CD82 could be an attractive molecular target to eradicate LSCs.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Gene Expression Regulation, Neoplastic , Kangai-1 Protein/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , ADP-ribosyl Cyclase 1/genetics , Animals , Antigens, CD34/genetics , Blotting, Western , Bone Marrow Transplantation , Cell Adhesion , Cell Movement , Cell Proliferation , Flow Cytometry , Homeodomain Proteins/physiology , Humans , Immunoenzyme Techniques , Kangai-1 Protein/antagonists & inhibitors , Kangai-1 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Neoplastic Stem Cells/metabolism , Peptide Fragments/analysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured
20.
Leuk Res ; 37(1): 76-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23102702

ABSTRACT

Shugoshin 1 (SGOL1), a centromeric protein, plays an important role in mitosis. This study explored the levels of SGOL1 in hematological malignancies and found that SGOL1 was aberrantly expressed in various human leukemia cell lines (n=10, e.g., HL60, U937, MOLM-13, K562, EOL-1, etc.) and freshly isolated leukemia cells from individuals with acute myelogenous leukemia (AML, n=43, p<0.001) compared with bone marrow mononuclear cells isolated from healthy volunteers (n=9), as measured by real-time RT-PCR. Forced expression of SGOL1 in hematopoietic stem/progenitor cells (HSPCs) significantly increased colony numbers for CFU-M and CFU-GM compared with control vector transduced infected HSPCs, suggesting that SGOL1 might act as an oncogene in hematopoietic cells. In addition, we found that repression of SGOL1 by small interfering RNA (siRNA) slowed the proliferation of NB4, EOL-1 and U937 cells compared with the control siRNA transfected cells, in parallel with the appearance of precocious dissociation of centromeric cohesion and separation of sister chromatids in these cells. Furthermore, we found that repression of SGOL1 by siRNA accumulated EOL-1 and U937 cells in the G2/M phase of the cell cycle, in conjunction with up-regulation of the spindle checkpoint protein BubR1, followed by apoptosis via caspase pathways. Thus, SGOL1 might be a promising molecular target for treating individuals with AML.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Leukemia/therapy , RNA, Small Interfering/genetics , Apoptosis , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Division , Cell Line, Tumor , G2 Phase , Humans , Leukemia/pathology , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...