Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biomol Ther (Seoul) ; 27(2): 201-209, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30419633

ABSTRACT

Mixed lineage leukemia proteins (MLL) are the key histone lysine methyltransferases that regulate expression of diverse genes. Aberrant activation of MLL promotes leukemia as well as solid tumors in humans, highlighting the urgent need for the development of an MLL inhibitor. We screened and isolated MLL1-binding ssRNAs using SELEX (Systemic Evolution of Ligands by Exponential enrichment) technology. When sequences in sub-libraries were obtained using next-generation sequencing (NGS), the most enriched aptamers-APT1 and APT2-represented about 30% and 26% of sub-library populations, respectively. Motif analysis of the top 50 sequences provided a highly conserved sequence: 5΄-A[A/C][C/G][G/U][U/A]ACAGAGGG[U/A]GG[A/C] GAGUGGGU-3΄. APT1, APT2, and APT5 embracing this motif generated secondary structures with similar topological characteristics. We found that APT1 and APT2 have a good binding activity and the analysis using mutated aptamer variants showed that the site information in the central region was critical for binding. In vitro enzyme activity assay showed that APT1 and APT2 had MLL1 inhibitory activity. Three-dimensional structure prediction of APT1-MLL1 complex indicates multiple weak interactions formed between MLL1 SET domain and APT1. Our study confirmed that NGS-assisted SELEX is an efficient tool for aptamer screening and that aptamers could be useful in diagnosis and treatment of MLL1-mediated diseases.

2.
Bioorg Med Chem Lett ; 27(3): 496-500, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28043794

ABSTRACT

Acute myeloid leukemia (AML) is a clonal disorder of hematopoietic progenitor cell. In AML, a mutation in FLT3 is commonly occurs and is associated with poor prognosis. We have previously reported that thieno[2,3-d]pyrimidine derivative compound 1 exhibited better antiproliferative activity against MV4-11 cells which harbor mutant FLT3 than AC220, which is a well-known FLT3 inhibitor, and has good microsomal stability. However, compound 1 had poor solubility. We then carried out further structural modification at the C2 and the C6 positions of thieno[2,3-d]pyrimidine scaffold. Compound 13b, which possesses a thiazole moiety at the C2 position, exhibited better antiproliferative activity than compound 1 and showed increased solubility and moderate microsomal stability. These results indicate that compound 13b could be a promising potential FLT inhibitor for AML chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Solubility , Structure-Activity Relationship , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
3.
Oncotarget ; 6(27): 24047-60, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26090722

ABSTRACT

The growth factors derived from the microenvironment create an environment conducive to tumor growth and survival. HGF deprivation using neutralizing antibody enhanced chemosensitivity in colorectal cancer cells (CRC). We determined secreted HGF in fibroblast conditioned medium (CM). Combination treatment of anti-HGF antibody and irinotecan (CPT-11) directly enhanced CPT-11 sensitivity in CRC. We generated xenograft in NOD/SCID mice inoculating HCT-116 human colorectal cancer cells subcutaneously with or without fibroblast. We found that the combination of CPT-11 and anti-HGF antibody induced marked suppression of tumor development. These results suggest that HGF produced by fibroblast induce CPT-11 resistance, and that anti-HGF antibody abrogate such resistance in vivo. fibroblast-derived HGF is important determinant of chemoresistance. Anti-HGF monoclonal antibody treatment confirmed the importance of this growth factor for chemoresistance in CRC. These results present new options toward the early diagnosis of chemoresistance and suggest novel combinations of chemotherapy and anti-HGF agents to prevent or significantly delay the onset of therapy resistance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Hepatocyte Growth Factor/immunology , Animals , Antibodies, Monoclonal/immunology , Apoptosis/physiology , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation , Culture Media, Conditioned/pharmacology , Enzyme Activation , Fibroblasts/metabolism , HCT116 Cells , HT29 Cells , Humans , Irinotecan , Lung Neoplasms/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-met/metabolism
4.
Oncol Rep ; 33(4): 1947-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672292

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) have been suggested as the potential new class of preventive or therapeutic antitumor agents. The aim of the present study was to evaluate the antitumor activity of the novel NSAID, CG100649. CG100649 is a novel NSAID dual inhibitor for COX-2 and carbonic anhydrase (CA)-I/-II. In the present study, we investigated the alternative mechanism by which CG100649 mediated suppression of the colon cancer growth and development. The anchorage­dependent and -independent clonogenic assay showed that CG100649 inhibited the clonogenicity of human colon cancer cells. The flow cytometric analysis showed that CG100649 induced the G2/M cell cycle arrest in colon cancer cells. Animal studies showed that CG100649 inhibited the tumor growth in colon cancer xenograft in nude mice. Furthermore, quantitative PCR and FACS analysis demonstrated that CG100649 upregulated the expression of TNF-related apoptosis-inducing ligand (TRAIL) receptors (DR4 and DR5) but decreased the expression of decoy receptors (DcR1 and DcR2) in colon cancer cells. The results showed that CG100649 treatment sensitized TRAIL­mediated growth suppression and apoptotic cell death. The combination treatment resulted in significant repression of the intestinal polyp formation in APCmin/+ mice. Our data clearly demonstrated that CG100649 contains preventive and therapeutic activity for colon cancer. The present study may be useful for identification of the potential benefit of the NSAID CG100649, for the achievement of a better treatment response in colon cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Furans/therapeutic use , Lung Neoplasms/drug therapy , Sulfonamides/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Synergism , Female , Furans/administration & dosage , Furans/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Intestinal Polyps/prevention & control , Intestine, Small/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor, Member 10c/biosynthesis , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Specific Pathogen-Free Organisms , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
5.
Eur J Cancer Prev ; 23(4): 277-85, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24871559

ABSTRACT

Histone deacetylase inhibitors (HDACi) have been reported to have potent chemopreventive activity because of their effects on the inhibition of cell growth and apoptosis in human cancer cell lines. In the present study, we investigated the apoptotic effect of a novel HDACi, Ky2, and its molecular mechanism in MDA-MB-231 human breast cancer cells in vitro. The chemopreventive effects of Ky2 in MDA-MB-231 cells were evaluated using the MTS assay, anchorage-independent cell transformation assay, DAPI staining, western blot analysis, reverse transcriptase-PCR, and small interfering RNA. Ky2 enhanced histone acetylation and decreased cell viability. Ky2 induced apoptosis evidenced by nuclear condensation and fragmentation, the accumulation of sub-G1 phase, and caspase-dependent PARP cleavage. In addition, Ky2 released cytochrome c from mitochondria to cytosol through the regulation of mitochondria-related proteins (Bid, Bim, and Bcl-xL). Ky2 markedly decreased the level of Sp1 protein expression through both the decrease of Sp1 mRNA level and proteasome-dependent protein degradation. Interestingly, the apoptotic effect of Ky2 is more potent than SAHA, a well-known HDACi. Furthermore, the knockdown of Sp1 protein by Sp1-specific inhibitor, mithramycin A, and siRNA resulted in the alteration of truncated Bid and Bim to induce apoptosis. Furthermore, Ky2 significantly decreased TPA-induced or EGF-induced neoplastic cell transformation in JB6 cells. Our results suggest that Ky2 may be a potential chemopreventive and chemotherapeutic agent by modulating Sp1 in human breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Peptides, Cyclic/pharmacology , RNA, Messenger/drug effects , Sp1 Transcription Factor/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chemoprevention , Cytochromes c/drug effects , Cytochromes c/metabolism , Drug Therapy , Female , Humans , RNA, Messenger/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
6.
Apoptosis ; 19(1): 179-90, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24085402

ABSTRACT

We previously reported that NSC126188 caused apoptosis of cancer cells by inducing expression of RhoB. We here present that NSC126188 induces apoptosis of prostate cancer PC-3 cells by inhibiting Akt/FoxO3 signaling, which mediates RhoB upregulation. The apoptosis and Akt dephosphorylation caused by NSC126188 was not substantially relieved by overexpressing wild-type Akt but was relieved by overexpressing constitutively active Akt (CA-Akt) or myristoylated Akt (myr-Akt). Furthermore, overexpression of CA-Akt or myr-Akt downregulated RhoB expression, indicating that RhoB expression is regulated by Akt signaling. Interestingly, membrane translocation of GFP-Akt by insulin exposure was abolished in the cells pretreated with NSC126188 suggesting that NSC126188 directly interfered with translocation of Akt to the plasma membrane. In addition, NSC126188 activated FoxO3a by dephosphorylating S253 via Akt inhibition. Activated FoxO3a translocated to the nucleus and increased transcription of RhoB and other target genes. PC-3 cells transiently overexpressing FoxO3a exhibited increased RhoB expression and apoptosis in response to NSC126188. Conversely, FoxO3a knockdown reduced NSC126188-induced RhoB expression and cell death. These results suggest that RhoB may be a target gene of FoxO3a and is regulated by Akt signaling. Taken together, NSC126188 induces apoptosis of PC-3 cells by interfering with membrane recruitment of Akt, resulting in Akt dephosphorylation and FoxO3a activation, which leads to transcription of RhoB.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Transcription Factors/metabolism , Piperazines/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , rhoB GTP-Binding Protein/genetics , Apoptosis , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Down-Regulation/drug effects , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Transcription, Genetic/drug effects , rhoB GTP-Binding Protein/metabolism
7.
Cancer Chemother Pharmacol ; 72(6): 1315-24, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24121479

ABSTRACT

PURPOSE: We investigated the action mechanism of a novel anticancer compound, KR28 (1-allyl-4-dodecanoyl-1-ethyl-piperazin-1-ium; bromide), to induce apoptosis of human prostate carcinoma PC-3 cells. METHODS: To explore an apoptotic signaling of KR28, we used ROS assay, SRB assay, flow cytometry analysis, reporter assay, xenograft assay, Western blotting, and RT-PCR analysis. RESULTS: The growth inhibitory action of KR28 is cell line specific, impeding the growth of prostate carcinoma PC-3 and stomach carcinoma NUGC-3 cells. KR28 showed strong antitumor activity in PC-3 mouse xenograft model. KR28 increased ROS production, leading to nuclear c-Abl expression, which in turn activated p38 mitogen-activated protein kinase (MAPK) to enhance the expression of RhoB, an apoptosis inducer. The KR28-induced apoptosis was abrogated by the ROS scavenger N-acetylcysteine and by knockdown of c-Abl, p38 MAPK, or ATF2. Moreover, the p300 binding site and two CCAAT boxes in the RhoB promoter appear to be involved in ROS-mediated RhoB expression in the presence of KR28. CONCLUSION: The antitumor agent KR28 induces apoptosis of PC-3 cells by ROS-mediated RhoB expression via c-Abl upregulation and activation of p38 MAPK/ATF-2.


Subject(s)
Allyl Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Piperazines/pharmacology , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-abl/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism , rhoB GTP-Binding Protein/genetics
8.
BMC Complement Altern Med ; 13: 194, 2013 Jul 27.
Article in English | MEDLINE | ID: mdl-23889969

ABSTRACT

BACKGROUND: Some of ginsenosides, root extracts from Panax ginseng, exert cytotoxicity against cancer cells through disruption of membrane subdomains called lipid rafts. Protopanaxadiol (PPD) exhibits the highest cytotoxic effect among 8 ginsenosides which we evaluated for anti-cancer activity. We investigated if PPD disrupts lipid rafts in its cytotoxic effects and also the possible mechanisms. METHODS: Eight ginsenosides were evaluated using different cancer cells and cell viability assays. The potent ginsenoside, PPD was investigated for its roles in lipid raft disruption and downstream pathways to apoptosis of cancer cells. Anti-cancer effects of PPD was also investigated in vivo using mouse xenograft model. RESULTS: PPD consistently exerts its potent cytotoxicity in 2 cell survival assays using 5 different cancer cell lines. PPD disrupts lipid rafts in different ways from methyl-ß-cyclodextrin (MßCD) depleting cholesterol out of the subdomains, since lipid raft proteins were differentially modulated by the saponin. During disruption of lipid rafts, PPD activated neutral sphingomyelinase 2 (nSMase 2) hydrolyzing membrane sphingomyelins into pro-apoptotic intracellular ceramides. Furthermore, PPD demonstrated its anti-cancer activities against K562 tumor cells in mouse xenograft model, confirming its potential as an adjunct or chemotherapeutic agent by itself in vivo. CONCLUSIONS: This study demonstrates that neutral sphingomyelinase 2 is responsible for the cytotoxicity of PPD through production of apoptotic ceramides from membrane sphingomyelins. Thus neutral sphingomyelinase 2 and its relevant mechanisms may potentially be employed in cancer chemotherapies.


Subject(s)
Cytotoxins/administration & dosage , Ginsenosides/administration & dosage , Neoplasms/drug therapy , Panax/chemistry , Sapogenins/administration & dosage , Sphingomyelin Phosphodiesterase/metabolism , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasms/enzymology , Neoplasms/genetics , Sphingomyelin Phosphodiesterase/genetics
9.
Life Sci ; 90(9-10): 373-80, 2012 Feb 27.
Article in English | MEDLINE | ID: mdl-22227470

ABSTRACT

AIMS: We investigated whether cAMP-mediated protein kinase A(PKA) and Epac1/Rap1 pathways differentially affect brain tumor cell death using 4-(3-cyclopentyloxy-4-methoxyphenyl)-2-pyrrolidone(rolipram), specific phosphodiesterase type IV(PDE IV) inhibitor. MAIN METHODS: A172 and U87MG human glioblastoma cells were used. Percentage of cell survival was determined by MTT assay. PKA and Epac1/Rap1 activation was determined by western blotting and pull-down assay, respectively. Cell cycle and hypodiploid cell formation were assessed by flow cytometry analysis. KEY FINDINGS: Non-specific PDE inhibitors, isobutylmethylxanthine(IBMX) and theophylline reduce survival percentage of A172 and U87MG cells. The expression of PDE4A and PDE4B was detected in A172 and U87MG cells. Rolipram-treated A172 or U87MG cell survival was lower in the presence of forskolin, adenylate cyclase activator, than that in its absence. Co-treatment with rolipram and forskolin also enhanced CREB phosphorylation on serine 133 that was inhibited by H-89, PKA inhibitor and cAMP-responsive guanine nucleotide exchange factor 1(Epac1), a Rap GDP exchange factor-mediated Rap1 activity in A172 cells. When A172 cells were treated with cell-permeable dibutyryl-cAMP(dbcAMP), PKA activator or 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate(CPT), Epac1 activator, basal level of cell death was increased and cell cycle was arrested at the phase of G2/M. Rolipram-induced A172 cell death was also increased by the co-treatment with dbcAMP or CPT, but it was inhibited by the pre-treatment with H-89. SIGNIFICANCE: These findings demonstrate that PKA and Epac1/Rap1 pathways could cooperatively play a role in rolipram-induced brain tumor cell death. It suggests that rolipram might regulate glioblastoma cell density through dual pathways of PKA- and Epac1/Rap1-mediated cell death and cell cycle arrest.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Brain Neoplasms/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Glioblastoma/enzymology , Guanine Nucleotide Exchange Factors/metabolism , Phosphodiesterase Inhibitors/pharmacology , Rolipram/pharmacology , Telomere-Binding Proteins/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 4/analysis , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dose-Response Relationship, Drug , Humans , Isoquinolines/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Shelterin Complex , Signal Transduction/drug effects , Sulfonamides/pharmacology , Theophylline/pharmacology
10.
Tumour Biol ; 33(2): 363-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22238052

ABSTRACT

Promoter CpG island hypermethylation of tumor suppressor genes is a common hallmark of all human cancers. Many researchers have been looking for potential epigenetic therapeutic targets in cancer using gene expression profiling with DNA microarray approaches. Our recent genome-wide platform of CpG island hypermethylation and gene expression in colorectal cancer (CRC) cell lines revealed that FBN2 and TCERG1L gene silencing is associated with DNA hypermethylation of a CpG island in the promoter region. In this study, promoter DNA hypermethylation of FBN2 and TCERG1L in CRC occurs as an early and cancer-specific event in colorectal cancer. Both genes showed high frequency of methylation in colon cancer cell lines (>80% for both of genes), adenomas (77% for FBN2, 90% for TCERG1L, n = 39), and carcinomas (86% for FBN2, 99% for TCERG1L, n = 124). Bisulfite sequencing confirmed cancer-specific methylation of FBN2 and TCERG1L of promoters in colon cancer cell line and cancers but not in normal colon. Methylation of FBN2 and TCERG1L is accompanied by downregulation in cell lines and in primary tumors as described in the Oncomine™ website. Together, our results suggest that gene silencing of FBN2 and TCERG1L is associated with promoter DNA hypermethylation in CRC tumors and may be excellent biomarkers for the early detection of CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , DNA Methylation , Early Detection of Cancer/methods , Aged , Cell Line, Tumor , Colonic Neoplasms/metabolism , CpG Islands , DNA Primers/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic
11.
Eur J Med Chem ; 46(8): 3258-64, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21570750

ABSTRACT

To define the SAR, a series of novel N-arylsulfonylimidazolidinone derivatives were evaluated for their in vitro anticancer activity against five human tumor cell lines, including A549, COLO205, KATO III, K562, SK-OV-3 and murine leukemia (P288D1) cell line. Among them, N-(2-chloroacetyl)-6-(2-oxo-4-phenylimidazolidin-1-ylsulfonyl)-3,4-dihydroquinoline-1(2H)-carboxamide (4m) and N-cyclohexyl-6-(2-oxo-4-phenylimidazolidin-1-ylsulfonyl)-3,4-dihydroquinoline-1(2H)-carboxamide (4n) exhibited comparable in vitro anticancer activity to doxorubicin against A549, KATO III and K562 cell lines and gave superior xenographic results against NCI-H23 and SW620 cancer cell lines. Regarding the structure-activity relationship, two critical points were discovered; the steric congestion at 4-position of N-arylsulfonylimidazolidinone scaffold abolishes the activity and the bulkiness or hydrophobicity of acyl groups at 3,4-dihydroquinoline of 4, especially with carbamoyl moiety, enormously enhances the activity.


Subject(s)
Antineoplastic Agents/pharmacology , Arylsulfonates/pharmacology , Colonic Neoplasms/drug therapy , Imidazolidines/pharmacology , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Arylsulfonates/chemical synthesis , Arylsulfonates/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/pathology , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Female , Humans , Hydrophobic and Hydrophilic Interactions , Imidazolidines/chemical synthesis , Imidazolidines/chemistry , Lung Neoplasms/pathology , Mice , Mice, Nude , Molecular Conformation , Neoplasm Transplantation , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Structure-Activity Relationship
12.
J Urol ; 185(6): 2366-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21511279

ABSTRACT

PURPOSE: Acetylation of chromatin interacting proteins is central to the epigenetic regulation of gene expression. Various tumor suppressors are inactivated by abnormal epigenetic modification. A great deal of effort has been devoted to developing anticancer agents that reactivate silenced tumor suppressors by modulating chromatin structure. Studies show that histone deacetylase inhibitors can act as anticancer agents and several histone deacetylase inhibitors are currently in clinical trials. We noted that the tumor suppressor RUNX3 is inactivated by promoter hypermethylation in human bladder cancer. We investigated whether reactivation of RUNX3 could suppress bladder cancer development in an animal model. MATERIALS AND METHODS: We analyzed RUNX3 reactivation and protein stabilization by a mild inhibitor of class III histone deacetylases, nicotinamide, by immunoprecipitation and immunoblot. Mouse bladder tumor was induced by N-butyl-N-(4-hydroxybutyl) nitrosamine. The effect of nicotinamide on Runx3 methylation status and tumor growth was measured. RESULTS: Nicotinamide induced RUNX3 expression at the transcriptional and posttranslational levels in a carcinogen induced mouse bladder tumor model and in human bladder tumor xenografts. Nicotinamide effectively inhibited the growth and progression of bladder tumors without decreasing body weight. CONCLUSIONS: Results suggest that nicotinamide has preventive and therapeutic effects on tumorigenesis through multiple mechanisms of RUNX3 expression up-regulation.


Subject(s)
Core Binding Factor Alpha 3 Subunit/drug effects , Core Binding Factor Alpha 3 Subunit/physiology , E1A-Associated p300 Protein/drug effects , E1A-Associated p300 Protein/physiology , Niacinamide/pharmacology , Up-Regulation/drug effects , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Animals , Disease Models, Animal , Humans , Mice , Sirtuin 2/physiology , Transplantation, Heterologous
13.
J Immunotoxicol ; 8(1): 56-67, 2011.
Article in English | MEDLINE | ID: mdl-21288165

ABSTRACT

Here, we investigated whether titanium dioxide (TiO2) nanoparticles affect in vivo tumor growth through the modulation of mononuclear leukocytes. In vitro lymphocyte proliferation by lipopolysaccharide (LPS) or concanavalin A (ConA) was reduced by < 25 nm TiO2 with a dose-dependent manner. Similarly, TiO2 nanoparticles inhibited nitric oxide (NO) production from bone marrow-derived macrophages obtained from naïve mice. When mice were intraperitoneally (IP) injected with < 25 or < 100 nm TiO2 once a day for 7 days, total cell number of splenocytes was reduced in the spleen of TiO2 nanoparticle-exposed mice. Both CD4+ and CD8+ T-lymphocyte numbers were significantly decreased and B-lymphocyte development was retarded by host exposure to the TiO2 nanoparticles. LPS-stimulated spleen cell proliferation was significantly reduced by host exposure to < 25 or < 100 nm TiO2, but no changes were detected in ConA-stimulated spleen cell proliferation. Further, LPS-stimulated cytokine production by peritoneal macrophages and the percentage of NK1.1+ natural killer cells among splenocytes was reduced by the host exposures to the TiO2 nanoparticles. When mice were IP injected with TiO2 nanoparticles once a day for 28 days prior to the subcutaneous implantation of B16F10 melanoma cells, tumor growth was subsequently significantly increased. Collectively, these results show that TiO2 nanoparticles may damage the development and proliferation of B- and T-lymphocytes, reduce the activity of macrophages, and decrease natural killer (NK) cell population levels, outcomes that appear to lead to an increase in tumor growth in situ. These studies allow us to suggest that TiO2 nanoparticles might have the potential to enhance tumor growth through immunomodulation of B- and T-lymphocytes, macrophages, and NK cells.


Subject(s)
Melanoma, Experimental/chemically induced , Melanoma, Experimental/immunology , Nanoparticles/toxicity , Titanium/toxicity , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Macrophages/drug effects , Macrophages/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology
14.
Genes Genet Syst ; 86(4): 277-84, 2011.
Article in English | MEDLINE | ID: mdl-22214596

ABSTRACT

DYX1C1 is a candidate gene for developmental dyslexia and has three alternative pre-mRNA spliced forms in the human genome. One of the transcripts contains an HERV-H LTR that could affect the expression level of DYX1C1. We speculate that the HERV-H LTR integrated into the DYX1C1 locus in the catarrhine lineage after its divergence from the platyrrhine lineage. Reverse transcription-PCR of the HERV-H LTR-related transcript produced four alternative forms from several human tissues. All of alternative forms were also identified in various rhesus macaque tissues. Through sequencing analysis of various primate DNA samples, we found that a part of the HERV-H LTR sequence was duplicated within the DYX1C1 exon 9 only in catarrhines. However, the duplication event did not cause frameshift mutation of the DYX1C1 transcript. Taken together, this HERV-H LTR insertion into DYX1C1 has contributed to transcript diversification of DYX1C1 during primate evolution.


Subject(s)
Endogenous Retroviruses/genetics , Evolution, Molecular , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Virus Integration , Animals , Base Sequence , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Chromosomes, Mammalian/virology , Computational Biology , Cytoskeletal Proteins , Exons , Gene Duplication , Genetic Variation , Genome, Human , Humans , Macaca mulatta/genetics , Macaca mulatta/metabolism , Macaca mulatta/virology , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phylogeny , Platyrrhini/genetics , Platyrrhini/metabolism , Platyrrhini/virology , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Species Specificity , Terminal Repeat Sequences
15.
J Immunol ; 185(7): 3980-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20826751

ABSTRACT

Vitamin D(3) upregulated protein 1 (VDUP1) is a candidate tumor suppressor, the expression of which is dramatically reduced in various tumor tissues. In this study, we found that VDUP1 expression is suppressed during human hepatic carcinogenesis, and mice lacking VDUP1 are much more susceptible to diethylnitrosamine-induced hepatocarcinogenesis compared with wild type mice. VDUP1-deficient tumors proliferated significantly more than wild type tumors and had corresponding changes in the expression of key cell cycle regulatory proteins. In addition, the hepatomitogen-induced response was associated with a considerable increase in the release of TNF-α and subsequent enhancement of NF-κB activation in VDUP1-deficient mice. When cells were treated with TNF-α, the VDUP1 level was markedly reduced, concomitant with elevated NF-κB activation. Furthermore, the overexpression of VDUP1 resulted in the robust suppression of TNF-α-activated NF-κB activity via association with HDAC1 and HDAC3. These results indicate that VDUP1 negatively regulates hepatocarcinogenesis by suppressing TNF-α-induced NF-κB activation.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Liver Neoplasms/metabolism , NF-kappa B/metabolism , Thioredoxins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Electrophoretic Mobility Shift Assay , Enzyme Activation/physiology , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Mice, Knockout , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
16.
Biochem Pharmacol ; 80(7): 982-9, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20599784

ABSTRACT

Hypoxia-inducible factor HIF-1 is responsible for radiation resistance and poor prognosis in cancer therapy. As part of our drug discovery program, a novel HIF inhibitor, LW6, was identified as a small compound that inhibits the accumulation of HIF-1alpha. We found that LW6 decreased HIF-1alpha protein expression without affecting HIF-1beta expression. MG132, a proteasome inhibitor, protected HIF-1alpha from LW6-induced proteasomal degradation, indicating that LW6 affects the stability of the HIF-1alpha protein. We found that LW6 promoted the degradation of wild type HIF-1alpha, but not of a DM-HIF-1alpha with modifications of P402A and P564A, at hydroxylation sites in the oxygen-dependent degradation domain (ODDD). LW6 did not affect the activity of prolyl hydroxylase (PHD), but induced the expression of von Hippel-Lindau (VHL), which interacts with prolyl-hydroxylated HIF-1alpha for proteasomal degradation. In the presence of LW6, knockdown of VHL did not abolish HIF-1alpha protein accumulation, indicating that LW6 degraded HIF-1alpha via regulation of VHL expression. In mice carrying xenografts of human colon cancer HCT116 cells, LW6 demonstrated strong anti-tumor efficacy in vivo and caused a decrease in HIF-1alpha expression in frozen-tissue immunohistochemical staining. These data suggest that LW6 may be valuable in the development of a HIF-1alpha inhibitor for cancer treatment.


Subject(s)
Acetanilides/pharmacology , Adamantane/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1/antagonists & inhibitors , von Hippel-Lindau Disease/genetics , Adamantane/pharmacology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Cell Line , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Female , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Mice, Nude , Oxygen/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteins/genetics , Proteins/metabolism , Up-Regulation , von Hippel-Lindau Disease/metabolism
17.
Bioorg Med Chem Lett ; 20(15): 4620-3, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20579876

ABSTRACT

Chroman derivatives exhibited potent inhibitory activity of NF-kappaB. For SAR, the chroman scaffold was modified with an indoline moiety. A series of indoline-2-carboxylic acid N-(substituted)phenylamide derivatives were synthesized to explore their inhibitory activities of NF-kappaB and they were also evaluated for cytotoxicity against various cancer cell lines. Since intermediates with Boc showed outstanding results, various substituents in place of the Boc group were introduced additionally and these compounds were also evaluated for SAR.


Subject(s)
Antineoplastic Agents/chemistry , Benzylamines/chemistry , Indoles/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Benzylamines/chemical synthesis , Benzylamines/toxicity , Cell Line, Tumor , Chromans/chemistry , Drug Screening Assays, Antitumor , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Structure-Activity Relationship
18.
Arch Pharm Res ; 33(5): 663-7, 2010 May.
Article in English | MEDLINE | ID: mdl-20512462

ABSTRACT

A series of 2-alkyl-2,3-dihydro-1H-2,6-diazacyclopenta[b]anthracene-5,10-diones (4a-f) was synthesized and their in vitro cytotoxic activities were evaluated against six human cancer cell lines (HCT15, SK-OV-3, SNB19, A549, MCF7 and MCF7/ADR). They all appeared to be less potent than doxorubicin against all doxorubicin sensitive human cancer cell lines tested. However, these compounds retained considerable cytotoxic activity against the doxorubicin-resistant cell line MCF7/ADR, implying their therapeutic potential to treat doxorubicin-resistant tumors. The most active compound 4c was equipotent with doxorubicin against HCT15 cell line.


Subject(s)
Anthracenes/chemical synthesis , Anthracenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor/methods , Humans , Molecular Structure , Quantitative Structure-Activity Relationship
19.
J Natl Cancer Inst ; 102(6): 426-42, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20194889

ABSTRACT

BACKGROUND: Vascular endothelial growth factor A (VEGFA), a critical mediator of tumor angiogenesis, is a well-characterized target of hypoxia-inducible factor 1 (HIF-1). Murine arrest-defective protein 1A (mARD1A(225)) acetylates HIF-1alpha, triggering its degradation, and thus may play a role in decreased expression of VEGFA. METHODS: We generated Apc(Min/+)/mARD1A(225) transgenic mice and quantified growth of intestinal polyps. Human gastric MKN74 and murine melanoma B16F10 cells overexpressing mARD1A(225) were injected into mice, and tumor growth and metastasis were measured. VEGFA expression and microvessel density in tumors were assessed using immunohistochemistry. To evaluate the role of mARD1A(225) acetylation of Lys532 in HIF-1alpha, we injected B16F10-mARD1A(225) cell lines stably expressing mutant HIF-1alpha/K532R into mice and measured metastasis. All statistical tests were two-sided, and P values less than .05 were considered statistically significant. RESULTS: Apc(Min/+)/mARD1A(225) transgenic mice (n = 25) had statistically significantly fewer intestinal polyps than Apc(Min/+) mice (n = 21) (number of intestinal polyps per mouse: Apc(Min/+) mice vs Apc(Min/+)/mARD1A(225) transgenic mice, mean = 83.4 vs 38.0 polyps, difference = 45.4 polyps, 95% confidence interval [CI] = 41.8 to 48.6; P < .001). The growth and metastases of transplanted tumors were also statistically significantly reduced in mice injected with mARD1A(225)-overexpressing cells than in mice injected with control cells (P < .01). Moreover, overexpression of mARD1A(225) decreased VEGFA expression and microvessel density in tumor xenografts (P < .04) and Apc(Min/+) intestinal polyps (P = .001). Mutation of lysine 532 of HIF-1alpha in B16F10-mARD1A(225) cells prevented HIF-1alpha degradation and inhibited the antimetastatic effect of mARD1A(225) (P < .001). CONCLUSION: mARD1A(225) may be a novel upstream target that blocks VEGFA expression and tumor-related angiogenesis.


Subject(s)
Acetyltransferases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Acetylation , Animals , Blotting, Western , Cell Line, Tumor , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Immunoprecipitation , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lysine , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Microcirculation , Mutation , N-Terminal Acetyltransferase A , N-Terminal Acetyltransferase E , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Vascular Endothelial Growth Factor A/analysis
20.
Bioorg Med Chem Lett ; 20(3): 1008-12, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20045644

ABSTRACT

A series of DAG-lactones with polar 3-alkylidene substituents have been investigated as PKC-alpha ligands and antitumor agents. Extensive analysis of structure-activity relationships for the 3-alkylidene chain revealed that polar groups such as ether, hydroxyl, aldehyde, ester, acyloxy, and amido were tolerated with similar binding affinities and reduced lipophilicities compared to the corresponding unsubstituted alkylidene chain. Among the derivatives, compounds 5, 6 and 8 with an ether type of side chain showed high binding affinities in range of K(i)= 3-5 nM and excellent antitumor profiles, particularly against the colo205 colon cancer and the K562 leukemia cell lines.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Protein Kinase C/metabolism , Valerates/chemistry , Valerates/metabolism , 4-Butyrolactone/chemistry , 4-Butyrolactone/metabolism , HL-60 Cells , Humans , K562 Cells , Lactones/chemistry , Lactones/metabolism , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL
...