Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
2.
Oncotarget ; 9(61): 31820-31831, 2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30159125

ABSTRACT

Protein methyltransferase SUV39H2 was reported to methylate histone H2AX at lysine 134 and enhance the formation of phosphorylated H2AX (γ-H2AX), which causes chemoresistance of cancer cells. We found that a series of imidazo[1,2-a]pyridine compounds that we synthesized could inhibit SUV39H2 methyltransferase activity. One of the potent compounds, OTS193320, was further analyzed in in vitro studies. The compound decreased global histone H3 lysine 9 tri-methylation levels in breast cancer cells and triggered apoptotic cell death. Combination of OTS193320 with doxorubicin (DOX) resulted in reduction of γ-H2AX levels as well as cancer cell viability compared to a single agent OTS193320 or DOX. Further optimization of inhibitors and their in vivo analysis identified a compound, OTS186935, which revealed significant inhibition of tumor growth in mouse xenograft models using MDA-MB-231 breast cancer cells and A549 lung cancer cells without any detectable toxicity. Our results suggest that the SUV39H2 inhibitors sensitize cancer cells to DOX by reduction of γ-H2AX levels in cancer cells, and collectively demonstrate that SUV39H2 inhibition warrants further investigation as a novel anti-cancer therapy.

3.
Oncotarget ; 8(34): 55837-55847, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28915556

ABSTRACT

Accumulation of ß-catenin in the nucleus is a hallmark of activation of the Wnt/ß-catenin signaling pathway, which drives development of a large proportion of human cancers. However, the mechanism of ß-catenin nuclear translocation has not been well investigated. Here we report biological significance of SMYD2-mediated lysine 133 (K133) methylation of ß-catenin on its nuclear translocation. Knockdown of SMYD2 attenuates the nuclear localization of ß-catenin protein in human cancer cells. Consequently, transcriptional levels of well-known Wnt-signaling molecules, cMYC and CCND1, are significantly reduced. Substitution of lysine 133 to alanine in ß-catenin almost completely abolishes its nuclear localization. We also demonstrate the K133 methylation is critical for the interaction of ß-catenin with FOXM1. Furthermore, after treatment with a SMYD2 inhibitor, significant reduction of nuclear ß-catenin and subsequent induction of cancer cell death are observed. Accordingly, our results imply that ß-catenin methylation by SMYD2 promotes its nuclear translocation and activation of Wnt signaling.

4.
Cancer Med ; 6(7): 1665-1672, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28639750

ABSTRACT

HER2 is a receptor tyrosine kinase, which is amplified and overexpressed in a subset of human cancers including breast and gastric cancers, and is indicated in its involvement in progression of cancer. Although its specific ligand(s) has not been detected, HER2 homodimerization, which is critical for its activation, is considered to be dependent on its expression levels. Here, we demonstrate a significant role of HER2 methylation by protein lysine methyltransferase SMYD3 in HER2 homodimerization. We found that SMYD3 trimethylates HER2 protein at lysine 175. HER2 homodimerization was enhanced in the presence of SMYD3, and substitution of lysine 175 of HER2 with alanine (HER2-K175A) reduced the formation of HER2 homodimers. Furthermore, HER2-K175A revealed lower level of autophosphorylation than wild-type HER2. We also identified that knockdown of SMYD3 attenuated this autophosphorylation in breast cancer cells. Our results imply that SMYD3-mediated methylation of HER2 at Lysine 175 may regulate the formation of HER2 homodimer and subsequent autophosphorylation and suggest that the SMYD3-mediated methylation pathway seems to be a good target for development of novel anti-cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Protein Multimerization , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Microsatellite Repeats , Phosphorylation , Polymorphism, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/metabolism
5.
Sci Rep ; 7: 40664, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102297

ABSTRACT

While multiple post-translational modifications have been reported to regulate the function of epidermal growth factor receptor (EGFR), the effect of protein methylation on its function has not been well characterized. In this study, we show that WHSC1L1 mono-methylates lysine 721 in the tyrosine kinase domain of EGFR, and that this methylation leads to enhanced activation of its downstream ERK cascade without EGF stimulation. We also show that EGFR K721 mono-methylation not only affects the function of cytoplasmic EGFR, but also that of nuclear EGFR. WHSC1L1-mediated methylation of EGFR in the nucleus enhanced its interaction with PCNA in squamous cell carcinoma of the head and neck (SCCHN) cells and resulted in enhanced DNA synthesis and cell cycle progression. Overall, our study demonstrates the multifaceted oncogenic function of the protein lysine methyltransferase WHSC1L1 in SCCHN, which is mediated through direct non-histone methylation of the EGFR protein with effects both in its cytoplasmic and nuclear functions.


Subject(s)
ErbB Receptors/genetics , Head and Neck Neoplasms/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/metabolism , Antineoplastic Agents/pharmacology , Biomarkers , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Replication , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Gene Knockdown Techniques , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Immunohistochemistry , Lysine/metabolism , Methylation , Models, Biological , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Transport
6.
Cancer Sci ; 108(3): 488-496, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28075524

ABSTRACT

T-lymphokine-activated killer cell-originated protein kinase (TOPK) plays critical roles in cancer cell proliferation as well as maintenance of cancer stem cells (CSC). Small cell lung cancer (SCLC) has highly aggressive phenotype, reveals early spread to distant sites, and results in dismal prognosis with little effective treatment. In this study, we demonstrate that TOPK expression was highly upregulated in both SCLC cell lines and primary tumors. Similar to siRNA-mediated TOPK knockdown effects, treatment with a potent TOPK inhibitor, OTS514, effectively suppressed growth of SCLC cell lines (IC50 ; 0.4-42.6 nM) and led to their apoptotic cell death. TOPK inhibition caused cell morphologic changes in SCLC cells, elongation of intercellular bridges caused by cytokinesis defects or neuronal protrusions induced by neuronal differentiation in a subset of CSC-like SCLC cells. Treatment with OTS514 suppressed forkhead box protein M1 (FOXM1) activity, which was involved in stemness of CSC. Furthermore, OTS514 treatment reduced CD90-positive SCLC cells and showed higher cytotoxic effect against lung sphere-derived CSC-like SCLC cells. Collectively, our results suggest that targeting TOPK is a promising approach for SCLC therapy.


Subject(s)
Cell Proliferation/drug effects , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Quinolones/pharmacology , Small Cell Lung Carcinoma/pathology , Thiophenes/pharmacology , Cell Proliferation/genetics , Forkhead Box Protein M1/antagonists & inhibitors , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/pathology , RNA Interference , RNA, Small Interfering/genetics , Spheroids, Cellular/drug effects , Thy-1 Antigens/metabolism , Tumor Cells, Cultured
7.
Medchemcomm ; 8(1): 73-80, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-30108692

ABSTRACT

Cancer stem cells (CSCs) are indicated to play critical roles in drug resistance, recurrence, and metastasis of cancer. Although molecular targeted therapies have contributed to the improvement of cancer treatments by targeting vulnerable pathways indispensable to the proliferation and survival of cancer cells, no relevant therapeutic modalities targeting CSCs have been developed yet. This review focuses on MELK (maternal embryonic leucine zipper kinase), TOPK (T-lymphokine-activated killer cell-originated protein kinase), and TTK (tyrosine threonine kinase), which are over-expressed frequently in human cancers and play indispensable roles in the development and maintenance of cancer stem cells. In addition, we will discuss recently developed small molecules for these protein targets, which have shown remarkable anti-tumor efficacies in several preclinical studies.

8.
Oncotarget ; 7(46): 75023-75037, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27626683

ABSTRACT

AKT1 is a cytosolic serine/threonine kinase that is overexpressed in various types of cancer and has a central role in human tumorigenesis. Although it is known that AKT1 is post-translationally modified in various ways including phosphorylation and ubiquitination, methylation has not been reported so far. Here we demonstrate that the protein lysine methyltrasnferase SMYD3 methylates lysine 14 in the PH domain of AKT1 both in vitro and in vivo. Lysine 14-substituted AKT1 shows significantly lower levels of phosphorylation at threonine 308 than wild-type AKT1, and knockdown of SMYD3 as well as treatment with a SMYD3 inhibitor significantly attenuates this phosphorylation in cancer cells. Furthermore, substitution of lysine 14 diminishes the plasma membrane accumulation of AKT1, and cancer cells overexpressing lysine 14-substiuted AKT1 shows lower growth rate than those overexpressing wild-type AKT1. These results imply that SMYD3-mediated methylation of AKT1 at lysine 14 is essential for AKT1 activation and that SMYD3-mediated AKT1 methylation appears to be a good target for development of anti-cancer therapy.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Pleckstrin Homology Domains , Proto-Oncogene Proteins c-akt/metabolism , Cell Membrane/metabolism , Enzyme Activation , Gene Expression , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Neoplasms/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/chemistry , Signal Transduction , Tyrosine/metabolism
9.
Oncotarget ; 7(14): 17652-64, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26933922

ABSTRACT

T-lymphokine-activated killer cell-originated protein kinase (TOPK) and maternal embryonic leucine zipper kinase (MELK) have been reported to play critical roles in cancer cell proliferation and maintenance of stemness. In this study, we investigated possible roles of TOPK and MELK in kidney cancer cells and found their growth promotive effect as well as some feedback mechanism between these two molecules. Interestingly, the blockade of either of these two kinases effectively caused downregulation of forkhead box protein M1 (FOXM1) activity which is known as an oncogenic transcriptional factor in various types of cancer cells. Small molecular compound inhibitors against TOPK (OTS514) and MELK (OTS167) effectively suppressed the kidney cancer cell growth, and the combination of these two compounds additively worked and showed the very strong growth suppressive effect on kidney cancer cells. Collectively, our results suggest that both TOPK and MELK are promising molecular targets for kidney cancer treatment and that dual blockade of OTS514 and OTS167 may bring additive anti-tumor effects with low risk of side effects.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Forkhead Box Protein M1/biosynthesis , Forkhead Box Protein M1/genetics , Forkhead Box Protein M1/metabolism , Gene Knockdown Techniques , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Targeted Therapy , Phosphorylation , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Small Molecule Libraries/pharmacology , Transfection
10.
Oncotarget ; 7(14): 18171-82, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26918358

ABSTRACT

MELK is upregulated in various types of human cancer and is known to be associated with cancer progression, maintenance of stemness, and poor prognosis. OTS167, a MELK kinase inhibitor, shows potent growth-suppressive effect on human tumors in a xenograft model, but the detailed mode of action has not been fully elucidated. In this study, we demonstrate the molecular mechanism of action of MELK inhibitor OTS167 in a preclinical model. OTS167-treated cells caused morphological transformation, induced the differentiation markers, and reduced stem-cell marker expression. Furthermore, we identified DEPDC1, known as an oncogene, as an additional downstream molecule of the MELK signaling pathway. MELK enhanced DEPDC1 phosphorylation and its stability. The expression of MELK and downstream molecules was decreased in OTS167-treated xenograft tumor tissues, which revealed central necrosis and significant growth suppression. Our data should further shed light on the mechanism of action how OTS167 suppresses tumor growth through the inhibition of the MELK signaling pathway and suggest the possibility of biomarkers for the assessment of clinical efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , GTPase-Activating Proteins/metabolism , Naphthyridines/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , A549 Cells , Animals , Biomarkers, Tumor , COS Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(12): 13621-33, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26871945

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK), that plays a critical role in maintenance of cancer stem cells (CSCs), is predominantly expressed in various types of human cancer including small cell lung cancer (SCLC). SCLC usually acquires resistance to anti-cancer drugs and portends dismal prognosis. We have delineated roles of MELK in development/progression of SCLC and examined anti-tumor efficacy of OTS167, a highly potent MELK inhibitor, against SCLC. MELK expression was highly upregulated in both SCLC cell lines and primary tumors. siRNA-mediated MELK knockdown induced significant growth inhibition in SCLC cell lines. Concordantly, treatment with OTS167 exhibited strong cytotoxicity against eleven SCLC cell lines with IC50 of < 10 nM. As similar to siRNA knockdown, OTS167 treatment induced cytokinetic defects with intercellular bridges, and in some cell lines we observed formation of neuronal protrusions accompanied with increase of a neuronal differentiation marker (CD56), indicating that the compound induced differentiation of cancer cells to neuron-like cells. Furthermore, the MELK inhibition decreased its downstream FOXM1 activity and Akt expression in SCLC cells, and led to apoptotic cell death. OTS167 appeared to be more effective to CSCs as measured by the sphere formation assay, thus MELK inhibition might become a promising treatment modality for SCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Box Protein M1/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Small Cell Lung Carcinoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Forkhead Box Protein M1/antagonists & inhibitors , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured
12.
Oncotarget ; 6(32): 33410-25, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26450903

ABSTRACT

Gain-of-function mutations of FLT3 (FLT3-ITD), comprises up to 30% of normal karyotype acute myeloid leukemia (AML) and is associated with an adverse prognosis. Current FLT3 kinase inhibitors have been tested extensively, but have not yet resulted in a survival benefit and novel therapies are awaited. Here we show that T-LAK cell-originated protein kinase (TOPK), a mitotic kinase highly expressed in and correlated with more aggressive phenotype in several types of cancer, is expressed in AML but not in normal CD34+ cells and that TOPK knockdown decreased cell viability and induced apoptosis. Treatment of AML cells with TOPK inhibitor (OTS514) resulted in a dose-dependent decrease in cell viability with lower IC50 in FLT3-mutated cells, including blasts obtained from patients relapsed after FLT3-inhibitor treatment. Using a MV4-11-engrafted mouse model, we found that mice treated with 7.5 mg/kg IV daily for 3 weeks survived significantly longer than vehicle treated mice (median survival 46 vs 29 days, P < 0.001). Importantly, we identified TOPK as a FLT3-ITD and CEBPA regulated kinase, and that modulating TOPK expression or activity resulted in significant decrease of FLT3 expression and CEBPA phosphorylation. Thus, targeting TOPK in FLT3-ITD AML represents a novel therapeutic approach for this adverse risk subset of AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Molecular Targeted Therapy , Mutation , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Tumor Cells, Cultured , U937 Cells
13.
Oncotarget ; 5(23): 12371-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25365263

ABSTRACT

Maternal embryonic leucine-zipper kinase (MELK), which was reported to be frequently up-regulated in various types of solid cancer, plays critical roles in formation and maintenance of cancer stem cells. However, little is known about the relevance of this kinase in hematologic malignancies. Here we report characterization of possible roles of MELK in acute myeloid leukemia (AML). MELK is expressed in AML cell lines and AML blasts with higher levels in less differentiated cells. MELK is frequently upregulated in AML with complex karyotypes and is associated with worse clinical outcome. MELK knockdown resulted in growth inhibition and apoptosis of leukemic cells. Hence, we investigated the potent anti-leukemia activity of OTS167, a small molecule MELK kinase inhibitor, in AML, and found that the compound induced cell differentiation and apoptosis as well as decreased migration of AML cells. MELK expression was positively correlated with the expression of FOXM1 as well as its downstream target genes. Furthermore, MELK inhibition resulted in downregulation of FOXM1 activity and the expression of its downstream targets. Taken together, and given that OTS167 is undergoing a phase I clinical trial in solid cancer, our study warrants clinical evaluation of this compound as a novel targeted therapy for AML patients.


Subject(s)
Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/metabolism , Protein Serine-Threonine Kinases/metabolism , Adolescent , Adult , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Transfection , Young Adult
14.
Sci Transl Med ; 6(259): 259ra145, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25338756

ABSTRACT

TOPK (T-lymphokine-activated killer cell-originated protein kinase) is highly and frequently transactivated in various cancer tissues, including lung and triple-negative breast cancers, and plays an indispensable role in the mitosis of cancer cells. We report the development of a potent TOPK inhibitor, OTS964 {(R)-9-(4-(1-(dimethylamino)propan-2-yl)phenyl)-8-hydroxy-6-methylthieno[2,3-c]quinolin-4(5H)-one}, which inhibits TOPK kinase activity with high affinity and selectivity. Similar to the knockdown effect of TOPK small interfering RNAs (siRNAs), this inhibitor causes a cytokinesis defect and the subsequent apoptosis of cancer cells in vitro as well as in xenograft models of human lung cancer. Although administration of the free compound induced hematopoietic adverse reactions (leukocytopenia associated with thrombocytosis), the drug delivered in a liposomal formulation effectively caused complete regression of transplanted tumors without showing any adverse reactions in mice. Our results suggest that the inhibition of TOPK activity may be a viable therapeutic option for the treatment of various human cancers.


Subject(s)
Cytokinesis , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokinesis/drug effects , Humans , Liposomes/chemistry , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Prognosis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Remission Induction , Treatment Outcome
15.
Oncotarget ; 3(12): 1629-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23283305

ABSTRACT

We previously reported MELK (maternal embryonic leucine zipper kinase) as a novel therapeutic target for breast cancer. MELK was also reported to be highly upregulated in multiple types of human cancer. It was implied to play indispensable roles in cancer cell survival and indicated its involvement in the maintenance of tumor-initiating cells. We conducted a high-throughput screening of a compound library followed by structure-activity relationship studies, and successfully obtained a highly potent MELK inhibitor OTSSP167 with IC50 of 0.41 nM. OTSSP167 inhibited the phosphorylation of PSMA1 (proteasome subunit alpha type 1) and DBNL (drebrin-like), which we identified as novel MELK substrates and are important for stem-cell characteristics and invasiveness. The compound suppressed mammosphere formation of breast cancer cells and exhibited significant tumor growth suppression in xenograft studies using breast, lung, prostate, and pancreas cancer cell lines in mice by both intravenous and oral administration. This MELK inhibitor should be a promising compound possibly to suppress the growth of tumor-initiating cells and be applied for treatment of a wide range of human cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell Proliferation/drug effects , Molecular Targeted Therapy , Naphthyridines/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Female , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Injections, Intravenous , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/metabolism , Molecular Structure , NIH 3T3 Cells , Naphthyridines/chemistry , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Small Molecule Libraries , Structure-Activity Relationship , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src Homology Domains
16.
J Med Chem ; 51(24): 7705-16, 2008 Dec 25.
Article in English | MEDLINE | ID: mdl-19032081

ABSTRACT

In silico screening is routinely used in the drug discovery process to predict whether each molecule in a database has a function of interest, such as inhibitory activity for a target protein. However, drugs generally have multiple functions including adverse effects. In order to obtain small molecules with desirable physiological effects, it is useful to simultaneously predict as many functions as possible. We employed Support Vector Machine to build classification models for 125 molecular functions, derived from the MDDR database, which showed higher kappa statistics (0.775 on average) than those of predictions by Tanimoto similarity (0.708). By analyzing the patterns of the predicted values (functional profiles) of 871 marketed drugs, we demonstrated its applications to indication discovery, clustering of drugs, and detection of molecular actions related to adverse effects. The results showed that functional profiling can be a useful tool for identifying the multifunctionality or adverse effects of small molecules.


Subject(s)
Technology, Pharmaceutical/methods , Algorithms , Chemistry, Pharmaceutical/methods , Cluster Analysis , Computer Simulation , Drug Design , Drug Discovery , Humans , Models, Statistical , Molecular Structure , Reproducibility of Results , Software , User-Computer Interface
17.
PLoS One ; 3(9): e3235, 2008 Sep 18.
Article in English | MEDLINE | ID: mdl-18800165

ABSTRACT

Recent in vitro and in vivo studies suggest that destabilized proteins with defective folding induce aggregation and toxicity in protein-misfolding diseases. One such unstable protein state is called amyloid oligomer, a precursor of fully aggregated forms of amyloid. Detection of various amyloid oligomers with A11, an anti-amyloid oligomer conformation-specific antibody, revealed that the amyloid oligomer represents a generic conformation and suggested that toxic beta-aggregation processes possess a common mechanism. By using A11 antibody as a probe in combination with mass spectrometric analysis, we identified GroEL in bacterial lysates as a protein that may potentially have an amyloid oligomer conformation. Surprisingly, A11 reacted not only with purified GroEL but also with several purified heat shock proteins, including human Hsp27, 40, 70, 90; yeast Hsp104; and bovine Hsc70. The native folds of A11-reactive proteins in purified samples were characterized by their anti-beta-aggregation activity in terms of both functionality and in contrast to the beta-aggregation promoting activity of misfolded pathogenic amyloid oligomers. The conformation-dependent binding of A11 with natively folded Hsp27 was supported by the concurrent loss of A11 reactivity and anti-beta-aggregation activity of heat-treated Hsp27 samples. Moreover, we observed consistent anti-beta-aggregation activity not only by chaperones containing an amyloid oligomer conformation but also by several A11-immunoreactive non-chaperone proteins. From these results, we suggest that the amyloid oligomer conformation is present in a group of natively folded proteins. The inhibitory effects of A11 antibody on both GroEL/ES-assisted luciferase refolding and Hsp70-mediated decelerated nucleation of Abeta aggregation suggested that the A11-binding sites on these chaperones might be functionally important. Finally, we employed a computational approach to uncover possible A11-binding sites on these targets. Since the beta-sheet edge was a common structural motif having the most similar physicochemical properties in the A11-reactive proteins we analyzed, we propose that the beta-sheet edge in some natively folded amyloid oligomers is designed positively to prevent beta aggregation.


Subject(s)
Amyloid/chemistry , Animals , Binding Sites , Cattle , Chaperonin 60/chemistry , Epitopes , HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins , Humans , Models, Statistical , Molecular Chaperones , Molecular Conformation , Protein Conformation , Protein Denaturation , Protein Folding , Protein Structure, Secondary
18.
Proteins ; 72(1): 367-81, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18214952

ABSTRACT

Many drugs, even ones that are designed to act selectively on a target protein, bind unintended proteins. These unintended bindings can explain side effects or indicate additional mechanisms for a drug's medicinal properties. Structural similarity between binding sites is one of the reasons for binding to multiple targets. We developed a method for the structural alignment of atoms in the solvent-accessible surface of proteins that uses similarities in the local atomic environment, and carried out all-against-all structural comparisons for 48,347 potential ligand-binding regions from a nonredundant protein structure subset (nrPDB, provided by NCBI). The relationships between the similarity of ligand-binding regions and the similarity of the global structures of the proteins containing the binding regions were examined. We found 10,403 known ligand-binding region pairs whose structures were similar despite having different global folds. Of these, we detected 281 region pairs that had similar ligands with similar binding modes. These proteins are good examples of convergent evolution. In addition, we found a significant correlation between Z-score of structural similarity and true positive rate of "active" entries in the PubChem BioAssay database. Moreover, we confirmed the interaction between ibuprofen and a new target, porcine pancreatic elastase, by NMR experiment. Finally, we used this method to predict new drug-target protein interactions. We obtained 540 predictions for 105 drugs (e.g., captopril, lovastatin, flurbiprofen, metyrapone, and salicylic acid), and calculated the binding affinities using AutoDock simulation. The results of these structural comparisons are available at http://www.tsurumi.yokohama-cu.ac.jp/fold/database.html.


Subject(s)
Biochemistry/methods , Pharmaceutical Preparations/metabolism , Proteins/chemistry , Proteins/metabolism , Algorithms , Binding Sites , Biological Assay , Ligands , Magnetic Resonance Spectroscopy , Protein Structure, Secondary , Reproducibility of Results
19.
Microbes Infect ; 8(1): 10-5, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16153874

ABSTRACT

The three-dimensional (3-D) structure of human immunodeficiency virus type 2 (HIV-2) Vpr/Vpx was predicted by homology modeling based on the NMR structure of human immunodeficiency virus type 1 (HIV-1) Vpr. The three proteins similarly have three major amphipathic alpha-helices. In contrast to HIV-1 Vpr, Vpr/Vpx of HIV-2 have a long N-terminal loop and clustered prolines in the second half of the C-terminal loop. HIV-2 Vpx uniquely contains a long region between the second and third major helices, and bears several glycines in the first half of the C-terminal loop. Instead of the glycines, there is a group of hydrophilic amino acids and arginines in the corresponding regions of the two Vprs. To compare the cytopathogenic potentials of HIV-1 Vpr and HIV-2 Vpr/Vpx, we examined the production of luciferase as a marker of cell damage. We further analyzed the characteristics of cells transduced with vpr/vpx genes driven by an inducible promoter. The results obtained clearly show that structurally similar, but distinct, HIV Vpr/Vpx proteins are detrimental to target cells.


Subject(s)
Cytopathogenic Effect, Viral/physiology , Gene Products, vpr/chemistry , Gene Products, vpr/metabolism , HIV-1 , HIV-2 , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/metabolism , Amino Acid Sequence , Gene Expression Regulation , Gene Products, vpr/genetics , HIV-1/chemistry , HIV-1/pathogenicity , HIV-2/chemistry , HIV-2/pathogenicity , HeLa Cells , Humans , Molecular Sequence Data , Protein Conformation , Sequence Alignment , Viral Regulatory and Accessory Proteins/genetics , vpr Gene Products, Human Immunodeficiency Virus
20.
J Biol Chem ; 280(26): 24915-22, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15870068

ABSTRACT

Interleukin-13 (IL-13) possesses two types of receptor: the heterodimer, composed of the IL-13Ralpha1 chain (IL-13Ralpha1) and the IL-4Ralpha chain (IL-4Ralpha), transducing the IL-13 signals; and the IL-13Ralpha2 chain (IL-13Ralpha2), acting as a nonsignaling "decoy" receptor. Extracellular portions of both IL-13Ralpha1 and IL-13Ralpha2 are composed of three fibronectin type III domains, D1, D2, and D3, of which the last two comprise the cytokine receptor homology modules (CRHs), a common structure of the class I cytokine receptor superfamily. Thus far, there has been no information about the critical amino acids of the CRHs or the role of the D1 domains of IL-13Ralpha1 and IL-13Ralpha2 in binding to IL-13. In this study, we first built the homology modeling of the IL-13.hIL-13 receptor complexes and then predicted the amino acids involved in binding to IL-13. By incorporating mutations into these amino acids, we identified Tyr-207, Asp-271, Tyr-315, and Asp-318 in the CRH of human IL-13Ralpha2, and Leu-319 and Tyr-321 in the CRH of human IL-13Ralpha1, as critical residues for binding to IL-13. Tyr-315 in IL-13Ralpha2 and Leu-319 in IL-13Ralpha1 are positionally conserved hydrophobic amino acid residues. Furthermore, by using D1 domain-deleted mutants, we found that the D1 domain is needed for the expression of IL-13Ralpha2, but not IL-13Ralpha1, and that the D1 domain of IL-13Ralpha1 is important for binding to IL-13, but not to IL-4. These results provide the basis for a precise understanding of the interaction between IL-13 and its receptors.


Subject(s)
Interleukin-13/chemistry , Receptors, Interleukin/chemistry , Amino Acid Sequence , Binding Sites , Cytokines/metabolism , Dimerization , Dose-Response Relationship, Drug , Fibronectins/chemistry , Gene Deletion , Humans , Interleukin-13 Receptor alpha1 Subunit , Interleukin-4/metabolism , Kinetics , Luciferases/metabolism , Models, Biological , Models, Genetic , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, Interleukin-13 , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...