Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 538
Filter
1.
Eur J Med Chem ; 189: 112023, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31978781

ABSTRACT

Disruptor of Telomeric Silencing 1-Like (DOT1L), the sole histone H3 lysine 79 (H3K79) methyltransferase, is required for leukemogenic transformation in a subset of leukemias bearing chromosomal translocations of the Mixed Lineage Leukemia (MLL) gene, as well as other cancers. Thus, DOT1L is an attractive therapeutic target and discovery of small molecule inhibitors remain of high interest. Herein, we are presenting screening results for a unique focused library of 1200 nucleoside analogs originally produced under the aegis of the NIH Pilot Scale Library Program. The complete nucleoside set was screened virtually against DOT1L, resulting in 210 putative hits. In vitro screening of the virtual hits resulted in validation of 11 compounds as DOT1L inhibitors clustered into two distinct chemical classes, adenosine-based inhibitors and a new chemotype that lacks adenosine. Based on the developed DOT1L ligand binding model, a structure-based design strategy was applied and a second-generation of non-nucleoside DOT1L inhibitors was developed. Newly synthesized compound 25 was the most potent DOT1L inhibitor in the new series with an IC50 of 1.0 µM, showing 40-fold improvement in comparison with hit 9 and exhibiting reasonable on target effects in a DOT1L dependent murine cell line. These compounds represent novel chemical probes with a unique non-nucleoside scaffold that bind and compete with the SAM binding site of DOT1L, thus providing foundation for further medicinal chemistry efforts to develop more potent compounds.


Subject(s)
Bone Marrow/drug effects , Cell Proliferation , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia, Experimental/drug therapy , Nucleosides/pharmacology , Triazoles/pharmacology , Animals , Bone Marrow/enzymology , Computer Simulation , Enzyme Inhibitors/chemistry , Leukemia, Experimental/enzymology , Mice , Nucleosides/chemistry , Structure-Activity Relationship , Triazoles/chemistry
2.
Blood ; 127(17): 2131-43, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26864341

ABSTRACT

Leukemias expressing constitutively activated mutants of ABL1 tyrosine kinase (BCR-ABL1, TEL-ABL1, NUP214-ABL1) usually contain at least 1 normal ABL1 allele. Because oncogenic and normal ABL1 kinases may exert opposite effects on cell behavior, we examined the role of normal ABL1 in leukemias induced by oncogenic ABL1 kinases. BCR-ABL1-Abl1(-/-) cells generated highly aggressive chronic myeloid leukemia (CML)-blast phase-like disease in mice compared with less malignant CML-chronic phase-like disease from BCR-ABL1-Abl1(+/+) cells. Additionally, loss of ABL1 stimulated proliferation and expansion of BCR-ABL1 murine leukemia stem cells, arrested myeloid differentiation, inhibited genotoxic stress-induced apoptosis, and facilitated accumulation of chromosomal aberrations. Conversely, allosteric stimulation of ABL1 kinase activity enhanced the antileukemia effect of ABL1 tyrosine kinase inhibitors (imatinib and ponatinib) in human and murine leukemias expressing BCR-ABL1, TEL-ABL1, and NUP214-ABL1. Therefore, we postulate that normal ABL1 kinase behaves like a tumor suppressor and therapeutic target in leukemias expressing oncogenic forms of the kinase.


Subject(s)
Blast Crisis/genetics , Genes, Tumor Suppressor , Genes, abl , Leukemia, Experimental/genetics , Leukemia, Myeloid, Chronic-Phase/genetics , Oncogene Proteins v-abl/physiology , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins c-abl/physiology , Tumor Suppressor Proteins/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blast Crisis/drug therapy , Blast Crisis/enzymology , Blast Crisis/pathology , Cell Division/drug effects , Cell Line, Tumor , Cytostatic Agents/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Genomic Instability , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Imidazoles/pharmacology , Imidazoles/therapeutic use , Leukemia, Experimental/drug therapy , Leukemia, Experimental/enzymology , Leukemia, Experimental/pathology , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/enzymology , Leukemia, Myeloid, Chronic-Phase/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Oxidative Stress , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-abl/genetics , Pyridazines/pharmacology , Pyridazines/therapeutic use , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
3.
Ann Hematol ; 95(5): 783-91, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891877

ABSTRACT

The novel FMS-like tyrosine kinase 3 (FLT3)-N676K point mutation within the FLT3 kinase domain-1 was recently identified in 6 % of de novo acute myeloid leukemia (AML) patients with inv(16). Because FLT3-N676K was encountered almost exclusively in inv(16) AML, we investigated the transforming potential of FLT3-N676K, the cooperation between FLT3-N676K and core binding factor ß-smooth muscle myosin heavy chain (CBFß-SMMHC) (encoded by the inv(16) chimeric gene CBFB-MYH11) in inducing acute leukemia, and tested the sensitivity of FLT3-N676K-positive leukemic cells to FLT3 inhibitors. Retroviral expression of FLT3-N676K in myeloid 32D cells induced AML in syngeneic C3H/HeJ mice (n = 11/13, median latency 58 days), with a transforming activity similar to FLT3-internal tandem duplication (ITD) (n = 8/8), FLT3-TKD D835Y (n = 8/9), and FLT3-ITD-N676K (n = 9/9) mutations. Three out of 14 (21.4 %) C57BL/6J mice transplanted with FLT3-N676K-transduced primary hematopoietic progenitor cells developed acute leukemia (latency of 68, 77, and 273 days), while no hematological malignancy was observed in the control groups including FLT3-ITD. Moreover, co-expression of FLT3-N676K/CBFß-SMMHC did not promote acute leukemia in three independent experiments (n = 16). In comparison with FLT3-ITD, FLT3-N676K induced much higher activation of FLT3 and tended to trigger stronger phosphorylation of MAPK and AKT. Importantly, leukemic cells carrying the FLT3-N676K mutant in the absence of an ITD mutation were highly sensitive to FLT3 inhibitors AC220 and crenolanib, and crenolanib even retained activity against the AC220-resistant FLT3-ITD-N676K mutant. Taken together, the FLT3-N676K mutant is potent to transform murine hematopoietic stem/progenitor cells in vivo. This is the first report of acute leukemia induced by an activating FLT3 mutation in C57BL/6J mice. Moreover, further experiments investigating molecular mechanisms for leukemogenesis induced by FLT3-N676K mutation and clinical evaluation of FLT3 inhibitors in FLT3-N676K-positive AML seem warranted.


Subject(s)
Leukemia, Experimental/genetics , Mutation, Missense , Point Mutation , fms-Like Tyrosine Kinase 3/genetics , Amino Acid Substitution , Animals , Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Benzothiazoles/therapeutic use , Bone Marrow Transplantation , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Genetic Vectors , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/enzymology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplastic Stem Cells/transplantation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phenylurea Compounds/therapeutic use , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational/genetics , Radiation Chimera , Retroviridae , Tandem Repeat Sequences , Transgenes , Tumor Stem Cell Assay , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/physiology
4.
J Virol ; 88(18): 10680-95, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24991018

ABSTRACT

UNLABELLED: Mammalian genomes are replete with retrotransposable elements, including endogenous retroviruses. DNA methyltransferase 3-like (DNMT3L) is an epigenetic regulator expressed in prospermatogonia, growing oocytes, and embryonic stem (ES) cells. Here, we demonstrate that DNMT3L enhances the interaction of repressive epigenetic modifiers, including histone deacetylase 1 (HDAC1), SET domain, bifurcated 1 (SETDB1), DNA methyltransferase 3A (DNMT3A), and tripartite motif-containing protein 28 (TRIM28; also known as TIF1ß and KAP1) in ES cells and orchestrates retroviral silencing activity with TRIM28 through mechanisms including, but not limited to, de novo DNA methylation. Ectopic expression of DNMT3L in somatic cells causes methylation-independent retroviral silencing activity by recruitment of the TRIM28/HDAC1/SETDB1/DNMT3A/DNMT3L complex to newly integrated Moloney murine leukemia virus (Mo-MuLV) proviral DNA. Concurrent with this recruitment, we also observed the accumulation of histone H3 lysine 9 trimethylation (H3K9me3) and heterochromatin protein 1 gamma (HP1γ), as well as reduced H3K9 and H3K27 acetylation at Mo-MuLV proviral sequences. Ectopic expression of DNMT3L in late-passage mouse embryonic fibroblasts (MEFs) recruited cytoplasmically localized HDAC1 to the nucleus. The formation of this epigenetic modifying complex requires interaction of DNMT3L with DNMT3A as well as with histone H3. In fetal testes at embryonic day 17.5, endogenous DNMT3L also enhanced the binding among TRIM28, DNMT3A, SETDB1, and HDAC1. We propose that DNMT3L may be involved in initiating a cascade of repressive epigenetic modifications by assisting in the preparation of a chromatin context that further attracts DNMT3A-DNMT3L binding and installs longer-term DNA methylation marks at newly integrated retroviruses. IMPORTANCE: Almost half of the mammalian genome is composed of endogenous retroviruses and other retrotransposable elements that threaten genomic integrity. These elements are usually subject to epigenetic silencing. We discovered that two epigenetic regulators that lack enzymatic activity, DNA methyltransferase 3-like (DNMT3L) and tripartite motif-containing protein 28 (TRIM28), collaborate with each other to impose retroviral silencing. In addition to modulating de novo DNA methylation, we found that by interacting with TRIM28, DNMT3L can attract various enzymes to form a DNMT3L-induced repressive complex to remove active marks and add repressive marks to histone proteins. Collectively, these results reveal a novel and pivotal function of DNMT3L in shaping the chromatin modifications necessary for retroviral and retrotransposon silencing.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Silencing , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Moloney murine leukemia virus/physiology , Repressor Proteins/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Gene Expression , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Leukemia, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Moloney murine leukemia virus/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Repressor Proteins/genetics , Tripartite Motif-Containing Protein 28
5.
Phytomedicine ; 20(10): 874-82, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23611488

ABSTRACT

The aim of this study was to explore whether the ethanolic extract of Antrodia cinnamomea (EEAC), a medical mushroom form Taiwan, could affect the proliferation and migration of WEHI-3 cells in vitro and to explore the antitumor effects of EEAC in BALB/c mice engrafted with WEHI-3 cells. The results showed that EEAC inhibited the proliferation of WEHI-3 cells, resulting in the accumulation of cell in G0/G1 and G2/M phases, as determined by flow cytometry. Moreover, EEAC markedly reduced the migration of WEHI-3 cells, as determined by a transwell assay. Treatment of WEHI-3 cells with EEAC also decreased MMP-9 protein expression and enzyme activity. The protein levels of p-Akt, p-ERK1/2 were also decreased, whereas the expression of p21 and p27 was increased. Furthermore, in an in vivo model, EEAC treatment reduced the infiltration of WEHI-3 cells into the liver and spleens and decreased tumor growth. Other bioactive compounds, such as cordycepin and zhankuic acid A, have been demonstrated to reduce the expression of MMP-9, cyclin E, cyclin D1 and to increase the expression of p21, p27. This is the first study to investigate that the mechanisms by which EEAC reduce the proliferation and migration of WEHI-3 cells in vitro, as well as the ability of EEAC to reduced infiltration of WEHI-3 cells into the liver and spleen in vivo. The results suggest that EEAC may prove to be useful in future antileukemic therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Antrodia , Cell Movement/drug effects , Cell Proliferation/drug effects , Fruiting Bodies, Fungal/chemistry , Leukemia, Experimental/drug therapy , Leukemia, Experimental/pathology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Fruiting Bodies, Fungal/physiology , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Experimental/enzymology , Mice , Mice, Inbred BALB C , Xenograft Model Antitumor Assays
6.
J Pharmacol Exp Ther ; 343(3): 617-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22935731

ABSTRACT

ABT-348 [1-(4-(4-amino-7-(1-(2-hydroxyethyl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-3-yl)phenyl)-3-(3-fluorophenyl)urea] is a novel ATP-competitive multitargeted kinase inhibitor with nanomolar potency (IC(50)) for inhibiting binding and cellular autophosphorylation of Aurora B (7 and 13 nM), C (1 and 13 nM), and A (120 and 189 nM). Cellular activity against Aurora B is reflected by inhibition of phosphorylation of histone H3, induction of polyploidy, and inhibition of proliferation of a variety of leukemia, lymphoma, and solid tumor cell lines (IC(50) = 0.3-21 nM). In vivo inhibition of Aurora B was confirmed in an engrafted leukemia model by observing a decrease in phosphorylation of histone H3 that persisted in a dose-dependent manner for 8 h and correlated with plasma concentration of ABT-348. Evaluation of ABT-348 across a panel of 128 kinases revealed additional potent binding activity (K(i) < 30 nM) against vascular endothelial growth factor receptor (VEGFR)/platelet-derived growth factor receptor (PDGFR) families and the Src family of cytoplasmic tyrosine kinases. VEGFR/PDGFR binding activity correlated with inhibition of autophosphorylation in cells and inhibition of vascular endothelial growth factor (VEGF)-stimulated endothelial cell proliferation (IC(50) ≤ 0.3 nM). Evidence of on-target activity in vivo was provided by the potency for blocking VEGF-mediated vascular permeability and inducing plasma placental growth factor. Activity against the Src kinase family was evident in antiproliferative activity against BCR-ABL chronic myeloid leukemia cells and cells expressing the gleevec-resistant BCR-ABL T315I mutation. On the basis of its unique spectrum of activity, ABT-348 was evaluated and found effective in representative solid tumor [HT1080 and pancreatic carcinoma (MiaPaCa), tumor stasis] and hematological malignancy (RS4;11, regression) xenografts. These results provide the rationale for clinical assessment of ABT-348 as a therapeutic agent in the treatment of cancer.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacokinetics , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Histones/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Molecular Structure , NIH 3T3 Cells , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/therapeutic use , Time Factors , Xenograft Model Antitumor Assays
7.
Biochem J ; 441(3): 889-99, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22014333

ABSTRACT

Mena [mammalian Ena (Enabled)]/VASP (vasodilator-stimulated phosphoprotein) proteins are the homologues of Drosophila Ena. In Drosophila, Ena is a substrate of the tyrosine kinase DAbl (Drosophila Abl). However, the link between Abl and the Mena/VASP family is not fully understood in mammals. We previously reported that Abi-1 (Abl interactor 1) promotes phosphorylation of Mena and BCAP (B-cell adaptor for phosphoinositide 3-kinase) by bridging the interaction between c-Abl and the substrate. In the present study we have identified VASP, another member of the Mena/VASP family, as an Abi-1-bridged substrate of Abl. VASP is phosphorylated by Abl when Abi-1 is co-expressed. We also found that VASP interacted with Abi-1 both in vitro and in vivo. VASP was tyrosine-phosphorylated in Bcr-Abl-positive leukaemic cells in an Abi-1-dependent manner. Co-expression of c-Abl and Abi-1 or the phosphomimetic Y39D mutation in VASP resulted in less accumulation of VASP at focal adhesions. VASP Y39D had a reduced affinity to the proline-rich region of zyxin. Interestingly, overexpression of both phosphomimetic and unphosphorylated forms of VASP, but not wild-type VASP, impaired adhesion of K562 cells to fibronectin. These results suggest that the phosphorylation and dephosphorylation cycle of VASP by the Abi-1-bridged mechanism regulates association of VASP with focal adhesions, which may regulate adhesion of Bcr-Abl-transformed leukaemic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Focal Adhesions/metabolism , Leukemia, Experimental/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , CHO Cells , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Cytoskeletal Proteins , Focal Adhesions/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Fusion Proteins, bcr-abl/physiology , HEK293 Cells , Humans , K562 Cells , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Mice , Microfilament Proteins/genetics , NIH 3T3 Cells , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/physiology , Tyrosine/metabolism , Xenopus laevis
8.
Cancer Res ; 71(8): 3052-65, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21493594

ABSTRACT

PDK1 activates AKT suggesting that PDK1 inhibition might suppress tumor development. However, while PDK1 has been investigated intensively as an oncology target, selective inhibitors suitable for in vivo studies have remained elusive. In this study we present the results of in vivo PDK1 inhibition through a universally applicable RNAi approach for functional drug target validation in oncogenic pathway contexts. This approach, which relies on doxycycline-inducible shRNA expression from the Rosa26 locus, is ideal for functional studies of genes like PDK1 where constitutive mouse models lead to strong developmental phenotypes or embryonic lethality. We achieved more than 90% PDK1 knockdown in vivo, a level sufficient to impact physiological functions resulting in hyperinsulinemia and hyperglycemia. This phenotype was reversible on PDK1 reexpression. Unexpectedly, long-term PDK1 knockdown revealed a lack of potent antitumor efficacy in 3 different mouse models of PTEN-deficient cancer. Thus, despite efficient PDK1 knockdown, inhibition of the PI3K pathway was marginal suggesting that PDK1 was not a rate limiting factor. Ex vivo analysis of pharmacological inhibitors revealed that AKT and mTOR inhibitors undergoing clinical development are more effective than PDK1 inhibitors at blocking activated PI3K pathway signaling. Taken together our findings weaken the widely held expectation that PDK1 represents an appealing oncology target.


Subject(s)
Neoplasms, Experimental/enzymology , PTEN Phosphohydrolase/deficiency , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Gene Knockdown Techniques , Gene Silencing , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Male , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Oncogene Protein v-akt/antagonists & inhibitors , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/genetics , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference
9.
Cancer Res ; 70(24): 10234-42, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159644

ABSTRACT

Chimeric oncoproteins resulting from fusion of MLL to a wide variety of partnering proteins cause biologically distinctive and clinically aggressive acute leukemias. However, the mechanism of MLL-mediated leukemic transformation is not fully understood. Dot1, the only known histone H3 lysine 79 (H3K79) methyltransferase, has been shown to interact with multiple MLL fusion partners including AF9, ENL, AF10, and AF17. In this study, we utilize a conditional Dot1l deletion model to investigate the role of Dot1 in hematopoietic progenitor cell immortalization by MLL fusion proteins. Western blot and mass spectrometry show that Dot1-deficient cells are depleted of the global H3K79 methylation mark. We find that loss of Dot1 activity attenuates cell viability and colony formation potential of cells immortalized by MLL oncoproteins but not by the leukemic oncoprotein E2a-Pbx1. Although this effect is most pronounced for MLL-AF9, we find that Dot1 contributes to the viability of cells immortalized by other MLL oncoproteins that are not known to directly recruit Dot1. Cells immortalized by MLL fusions also show increased apoptosis, suggesting the involvement of Dot1 in survival pathways. In summary, our data point to a pivotal requirement for Dot1 in MLL fusion protein-mediated leukemogenesis and implicate Dot1 as a potential therapeutic target.


Subject(s)
Cell Transformation, Neoplastic/genetics , Methyltransferases/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Apoptosis/physiology , Cell Transformation, Neoplastic/metabolism , Hematopoietic Stem Cells/enzymology , Histone-Lysine N-Methyltransferase , Histones/metabolism , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Leukemia, Experimental/pathology , Lysine/metabolism , Methylation , Methyltransferases/antagonists & inhibitors , Methyltransferases/deficiency , Methyltransferases/genetics , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism
10.
J Immunol ; 185(6): 3305-12, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20693424

ABSTRACT

Indoleamine 2,3-dioxygenase, the L-tryptophan-degrading enzyme, plays a key role in the powerful immunomodulatory effects on several different types of cells. Because modulation of IDO activities after viral infection may have great impact on disease progression, we investigated the role of IDO following infection with LP-BM5 murine leukemia virus. We found suppressed BM5 provirus copies and increased type I IFNs in the spleen from IDO knockout (IDO(-/-)) and 1-methyl-D-L-tryptophan-treated mice compared with those from wild-type (WT) mice. Additionally, the number of plasmacytoid dendritic cells in IDO(-/-) mice was higher in the former than in the WT mice. In addition, neutralization of type I IFNs in IDO(-/-) mice resulted in an increase in LP-BM5 viral replication. Moreover, the survival rate of IDO(-/-) mice or 1-methyl-D-L-tryptophan-treated mice infected with LP-BM5 alone or with both Toxoplasma gondii and LP-BM5 was clearly greater than the survival rate of WT mice. To our knowledge, the present study is the first report to observe suppressed virus replication with upregulated type I IFN in IDO(-/-) mice, suggesting that modulation of the IDO pathway may be an effective strategy for treatment of virus infection.


Subject(s)
Down-Regulation/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Interferon Type I/biosynthesis , Leukemia Virus, Murine/immunology , Retroviridae Infections/enzymology , Retroviridae Infections/prevention & control , Up-Regulation/immunology , Virus Replication/immunology , Adaptive Immunity/genetics , Animals , Down-Regulation/genetics , Immunity, Innate/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon Type I/physiology , Leukemia Virus, Murine/growth & development , Leukemia, Experimental/enzymology , Leukemia, Experimental/immunology , Leukemia, Experimental/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Virus Infections/enzymology , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Up-Regulation/genetics
11.
J Clin Invest ; 119(12): 3774-86, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19907075

ABSTRACT

When used as therapy for hematopoietic malignancies, allogeneic BM transplantation (BMT) relies on the graft-versus-leukemia (GVL) effect to eradicate residual tumor cells through immunologic mechanisms. However, graft-versus-host disease (GVHD), which is initiated by alloreactive donor T cells that recognize mismatched major and/or minor histocompatibility antigens and cause severe damage to hematopoietic and epithelial tissues, is a potentially lethal complication of allogeneic BMT. To enhance the therapeutic potential of BMT, we sought to find therapeutic targets that could inhibit GVHD while preserving GVL and immune responses to infectious agents. We show here that T cell responses triggered in mice by either Listeria monocytogenes or administration of antigen and adjuvant were relatively well preserved in the absence of PKC isoform theta (PKCtheta), a key regulator of TCR signaling. In contrast, PKCtheta was required for alloreactivity and GVHD induction. Furthermore, absence of PKCtheta raised the threshold for T cell activation, which selectively affected alloresponses. Most importantly, PKCtheta-deficient T cells retained the ability to respond to virus infection and to induce GVL effect after BMT. These findings suggest PKCtheta is a potentially unique therapeutic target required for GVHD induction but not for GVL or protective responses to infectious agents.


Subject(s)
Graft vs Host Disease/enzymology , Graft vs Leukemia Effect/physiology , Isoenzymes/immunology , Leukemia, Experimental/enzymology , Leukemia, Experimental/immunology , Protein Kinase C/immunology , Retroviridae Infections/enzymology , Retroviridae Infections/immunology , Animals , Bone Marrow Transplantation/immunology , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , In Vitro Techniques , Isoantigens , Isoenzymes/deficiency , Isoenzymes/genetics , Listeria monocytogenes/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C-theta , Signal Transduction , T-Lymphocytes/immunology
12.
Exp Hematol ; 37(12): 1423-34, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19819294

ABSTRACT

OBJECTIVE: Animal models have provided evidence for the existence of leukemia stem cells (LSC). However, prospective isolation of human LSC from patients with acute myeloid leukemia (AML), as well as the assessment of their clinical significance, has remained a major challenge. MATERIALS AND METHODS: We have studied the functional characteristics of a subset of leukemia cells that expressed CD34 and high aldehyde dehydrogenase activity (ALDH(br)), which was freshly isolated from the mononuclear cells at the time of diagnosis from the marrow of 68 consecutive patients suffering from AML. RESULTS: The percentage of ALDH(br) cells ranged from 0.01% to 16.0% with a median of 0.5%. Compared to their counterparts with low aldehyde dehydrogenase activity from the same individual patients, the ALDH(br) population showed a significantly higher affinity to human mesenchymal stromal cells (n=12; p<0.01), a more than twofold higher proportion of slow-dividing and quiescent cells (n=4; p<0.05), higher numbers of long-term culture-initiating cell colonies in vitro (n=25; p<0.01), and an enhanced engraftment in the nonobese diabetic/severe combined immunodeficient mouse model (n=3; p<0.05). Above all, we found that the frequency of ALDH(br) cells correlated significantly with diminished survival probability (p=0.025) and with adverse cytogenetic factors (p<0.05). CONCLUSION: A small proportion of leukemia cells derived from the marrow of patients with AML were ALDH(br) and CD34(+). They demonstrated functional characteristics of LSC and high percentages of these cells among the leukemia cells correlated significantly with poor clinical outcome.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Leukemia, Myeloid/enzymology , Neoplastic Stem Cells/enzymology , Acute Disease , Adult , Aged , Aldehyde Dehydrogenase/genetics , Animals , Antigens, CD34/metabolism , Female , Flow Cytometry , Humans , Leukemia, Experimental/enzymology , Leukemia, Experimental/pathology , Leukemia, Myeloid/blood , Leukemia, Myeloid/pathology , Male , Mesoderm/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplastic Stem Cells/pathology , Prognosis , Stromal Cells/enzymology , Stromal Cells/pathology , Survival Analysis , Transplantation, Heterologous , Tumor Cells, Cultured , Young Adult
13.
Chembiochem ; 10(16): 2638-43, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19746508

ABSTRACT

The natural product syringolin A (SylA) is a potent proteasome inhibitor with promising anticancer activities. To further investigate its potential as a lead structure, selectivity profiling with cell lysates was performed. At therapeutic concentrations, a rhodamine-tagged SylA derivative selectively bound to the 20 S proteasome active sites without detectable off-target labelling. Additional profiling with lysates of wild-type and bortezomib-adapted leukaemic cell lines demonstrated the retention of this proteasome target and subsite selectivity as well as potency even in clinically relevant cell lines. Our studies, therefore, propose that further development of SylA might indeed result in an improved small molecule for the treatment of leukaemia.


Subject(s)
Boronic Acids/administration & dosage , Leukemia, Experimental/pathology , Peptides, Cyclic/pharmacology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/administration & dosage , Animals , Bortezomib , Cell Line, Tumor , Leukemia, Experimental/enzymology , Magnetic Resonance Spectroscopy , Mice , Spectrometry, Mass, Electrospray Ionization
14.
J Mol Biol ; 385(1): 65-78, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-18983852

ABSTRACT

Mammalian APOBEC molecules comprise a large family of cytidine deaminases with specificity for RNA and single-stranded DNA (ssDNA). APOBEC1s are invariably highly specific and edit a single residue in a cellular mRNA, while the cellular targets for APOBEC3s are not clearly established, although they may curtail the transposition of some retrotransposons. Two of the seven member human APOBEC3 enzymes strongly restrict human immunodeficiency virus type 1 in vitro and in vivo. We show here that ssDNA hyperediting of an infectious exogenous gammaretrovirus, the Friend-murine leukemia virus, by murine APOBEC1 and APOBEC3 deaminases occurs in vitro. Murine APOBEC1 was able to hyperdeaminate cytidine residues in murine leukemia virus genomic RNA as well. Analysis of the edited sites shows that the deamination in vivo was due to mouse APOBEC1 rather than APOBEC3. Furthermore, murine APOBEC1 is able to hyperedit its primary substrate in vivo, the apolipoprotein B mRNA, and a variety of heterologous RNAs. In short, murine APOBEC1 is a hypermutator of both RNA and ssDNA in vivo, which could exert occasional side effects upon overexpression.


Subject(s)
Cytidine Deaminase/metabolism , Leukemia Virus, Murine/metabolism , Mutation/genetics , RNA/genetics , APOBEC Deaminases , APOBEC-1 Deaminase , Animals , Animals, Newborn , Apolipoproteins B/genetics , Base Sequence , DNA, Complementary/genetics , Genome, Viral/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Hypoxanthine Phosphoribosyltransferase/metabolism , Leukemia Virus, Murine/genetics , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Mice , Molecular Sequence Data , Muscle Proteins/metabolism , NIH 3T3 Cells , Nucleic Acid Denaturation , Nucleotides , RNA Editing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae Infections/enzymology , Tumor Virus Infections/enzymology , Tumor Virus Infections/genetics
15.
Leukemia ; 23(3): 585-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18754030
16.
Cancer Sci ; 99(6): 1251-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18429956

ABSTRACT

Despite promising results from clinical studies of ABL kinase inhibitors, a challenging problem that remains is the T315I mutation against which neither nilotinib nor dasatinib show significant activity. In the present study, we investigated the activity of a novel Aurora kinase inhibitor, VE-465, against leukemia cells expressing wild-type BCR-ABL or the T315I mutant form of BCR-ABL. We observed a dose-dependent reduction in the level of BCR-ABL autophosphorylation in VE-465-treated cells. Exposure to the combination of VE-465 and imatinib exerted an enhanced apoptotic effect in K562 cells. Combined treatment with VE-465 and imatinib caused more attenuation of the levels of phospho-AKT and c-Myc in K562 cells. Further, the isobologram indicated the synergistic effect of simultaneous exposure to VE-465 and imatinib in K562 cells. To assess the in vivo efficacy of VE-465, athymic nude mice were injected intravenously with BaF3 cells expressing wild-type BCR-ABL or the T315I mutant form. The vehicle-treated mice died of a condition resembling acute leukemia by 28 days; however, nearly all mice treated with VE-465 (75 mg/kg, twice daily; intraperitoneally for 14 days) survived for more than 56 days. Histopathological analysis of vehicle-treated mice revealed infiltration of the spleen. In contrast, histopathological analysis of organs from VE-465-treated mice demonstrated normal tissue architecture. Taken together, the present study shows that VE-465 exhibits a desirable therapeutic index that can reduce the in vivo growth of T315I mutant form and wild-type BCR-ABL-expressing cells in an efficacious manner.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/metabolism , Leukemia, Experimental/drug therapy , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Aurora Kinases , Benzamides , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Therapy, Combination , Female , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Immunoblotting , Immunoprecipitation , K562 Cells , Leukemia, Experimental/enzymology , Leukemia, Experimental/metabolism , Mice , Mice, Nude , Phosphorylation/drug effects , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Survival Rate
17.
Leukemia ; 22(6): 1117-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18401417

ABSTRACT

Amplification of the NUP214-ABL1 oncogene can be detected in patients with T cell acute lymphoblastic leukemia (T-ALL). We screened 29 patients with T cell malignancies for the expression of NUP214-ABL1 by reverse transcription-polymerase chain reaction (RT-PCR). NUP214-ABL1 was detected in three (10%) patients. These results were confirmed by fluorescence in situ hybridization techniques. We also studied the activity of imatinib, nilotinib and dasatinib against the human NUP214-ABL1-positive cell lines PEER and BE-13. All three tyrosine kinase inhibitors decreased the viability of PEER and BE-13 cells, but nilotinib and dasatinib had >1-log lower IC(50) values than imatinib (P<0.001). In contrast, the NUP214-ABL-negative T-ALL cell line Jurkat, was remarkably resistant to tyrosine kinase inhibition. The inhibition of cellular proliferation was associated with time-dependent induction of apoptosis and inhibition of ABL, CrKL and STAT5 phosphorylation. Moreover, dasatinib was active in a NUP214-ABL1-positive leukemia xenograft murine model and in marrow lymphoblasts from a patient with NUP214-ABL1-positive T-ALL. On the basis of these results, ABL1 kinase inhibitors warrant clinical investigation in patients with NUP214-ABL1-positive T-cell malignancies.


Subject(s)
Leukemia, Experimental/drug therapy , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Adult , Animals , Apoptosis/drug effects , Benzamides , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dasatinib , Female , Humans , Imatinib Mesylate , In Situ Hybridization, Fluorescence , Leukemia, Experimental/enzymology , Leukemia, Experimental/genetics , Leukemia-Lymphoma, Adult T-Cell/enzymology , Leukemia-Lymphoma, Adult T-Cell/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Phosphorylation/drug effects , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism , Thiazoles/therapeutic use , Tumor Cells, Cultured
18.
J Virol ; 81(19): 10506-14, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17634228

ABSTRACT

The roles of cellular proteases in Moloney murine leukemia virus (MLV) infection were investigated using MLV particles pseudotyped with vesicular stomatitis virus (VSV) G glycoprotein as a control for effects on core MLV particles versus effects specific to Moloney MLV envelope protein (Env). The broad-spectrum inhibitors cathepsin inhibitor III and E-64d gave comparable dose-dependent inhibition of Moloney MLV Env and VSV G pseudotypes, suggesting that the decrease did not involve the envelope protein. Whereas, CA-074 Me gave a biphasic response that differentiated between Moloney MLV Env and VSV G at low concentrations, at which the drug is highly selective for cathepsin B, but was similar for both glycoproteins at higher concentrations, at which CA-074 Me inhibits other cathepsins. Moloney MLV infection was lower on cathepsin B knockout fibroblasts than wild-type cells, whereas VSV G infection was not reduced on the B-/- cells. Taken together, these results support the notion that cathepsin B acts at an envelope-dependent step while another cathepsin acts at an envelope-independent step, such as uncoating or viral-DNA synthesis. Virus binding was not affected by CA-074 Me, whereas syncytium induction was inhibited in a dose-dependent manner, consistent with cathepsin B involvement in membrane fusion. Western blot analysis revealed specific cathepsin B cleavage of SU in vitro, while TM and CA remained intact. Infection could be enhanced by preincubation of Moloney MLV with cathepsin B, consistent with SU cleavage potentiating infection. These data suggested that during infection of NIH 3T3 cells, endocytosis brings Moloney MLV to early lysosomes, where the virus encounters cellular proteases, including cathepsin B, that cleave SU.


Subject(s)
Cathepsin B/metabolism , Leukemia, Experimental/enzymology , Membrane Fusion , Moloney murine leukemia virus , Animals , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Endocytosis , Leucine/analogs & derivatives , Leucine/pharmacology , Lysosomes/enzymology , Lysosomes/virology , Membrane Fusion/drug effects , Mice , NIH 3T3 Cells
19.
Immunity ; 24(6): 671-672, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782023

ABSTRACT

AID is a cytidine deaminase essential for class switch recombination and somatic hypermutation during the humoral immune response. In this issue of Immunity, Gourzi et al. (2006) show that AID also plays a critical role in innate immunity to virally induced acute pro-B cell leukemia.


Subject(s)
Abelson murine leukemia virus/immunology , Cytidine Deaminase/physiology , Leukemia, Experimental/enzymology , Retroviridae Infections/enzymology , Tumor Virus Infections/enzymology , Animals , Cytidine Deaminase/genetics , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Mice , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology
20.
Immunity ; 24(6): 779-786, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782033

ABSTRACT

Activation-induced cytidine deaminase (AID) is specifically expressed in the germinal centers of lymphoid organs, where it initiates targeted hypermutation of variable regions of immunoglobulin genes in response to stimulation by antigen. Ectopic expression of AID, however, mediates generalized hypermutation in eukaryotes and prokaryotes. Here, we present evidence that AID is induced outside the germinal center in response to infection by the Abelson murine leukemia virus. The genotoxic activity of virally induced AID resulted in checkpoint kinase-1 (chk1) phosphorylation and ultimately restricted the proliferation of the infected cell. At the same time, it induced NKG2D ligand upregulation, which alerts the immune system to the presence of virally transformed cells. Hence, in addition to its known function in immunoglobulin diversification, AID is active in innate defense against a transforming retrovirus.


Subject(s)
Abelson murine leukemia virus/immunology , Cytidine Deaminase/physiology , Leukemia, Experimental/enzymology , Retroviridae Infections/enzymology , Tumor Virus Infections/enzymology , Animals , B-Lymphocytes/enzymology , Bone Marrow/enzymology , Bone Marrow/virology , Checkpoint Kinase 1 , Cytidine Deaminase/genetics , Death , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Ligands , Mice , Mice, Inbred Strains , NK Cell Lectin-Like Receptor Subfamily K , Phosphorylation , Protein Kinases/metabolism , Receptors, Immunologic/metabolism , Receptors, Natural Killer Cell , Retroviridae Infections/genetics , Retroviridae Infections/immunology , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL