Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell ; 156(4): 663-77, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24529372

ABSTRACT

Cancer is believed to arise primarily through accumulation of genetic mutations. Although induced pluripotent stem cell (iPSC) generation does not require changes in genomic sequence, iPSCs acquire unlimited growth potential, a characteristic shared with cancer cells. Here, we describe a murine system in which reprogramming factor expression in vivo can be controlled temporally with doxycycline (Dox). Notably, transient expression of reprogramming factors in vivo results in tumor development in various tissues consisting of undifferentiated dysplastic cells exhibiting global changes in DNA methylation patterns. The Dox-withdrawn tumors arising in the kidney share a number of characteristics with Wilms tumor, a common pediatric kidney cancer. We also demonstrate that iPSCs derived from Dox-withdrawn kidney tumor cells give rise to nonneoplastic kidney cells in mice, proving that they have not undergone irreversible genetic transformation. These findings suggest that epigenetic regulation associated with iPSC derivation may drive development of particular types of cancer.


Subject(s)
Cellular Reprogramming , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Animals , DNA Methylation , Doxycycline/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney Neoplasms/chemically induced , Mice , Mice, Transgenic , Transcription Factors/metabolism
2.
Anticancer Drugs ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39259581

ABSTRACT

Prostate cancer antigen-1/ALKBH3, a DNA/RNA demethylase of 3-methylcytosine, 1-methyladenine (1-meA), and 6-meA, was found in prostate cancer as an important prognostic factor. Additionally, 1-meA has been associated with other cancers. The ALKBH3 inhibitor HUHS015 was found to be effective against prostate cancer both in vitro and in vivo. Herein, we investigated the effect of HUHS015 in combination with drugs for prostate cancer approved in Japan (including bicalutamide, cisplatin, mitoxantrone, prednisolone, ifosfamide, tegafur/uracil, docetaxel, dacarbazine, and estramustine) by treating DU145 cells with around IC50 value concentrations of these drugs for 3 days. Additionally, the cells were observed for additional 9 days after drug removal. Combination treatment with dacarbazine, estramustine, tegafur/uracil, and HUHS015 showed a slight additive effect after 3 days. After drug washout of them and mitoxantrone, the combined effects and levels were enhanced and sustained, although the effects of each treatment alone declined. HUHS015 combined with cisplatin or docetaxel elicited synergistic and sustained effects. In vivo, combining HUHS015 and docetaxel, the first chemotherapeutic agent for castration-resistant prostate cancer, showed notable effects in the DU145 xenograft model. In conclusion, HUHS015 exhibited a synergistic effect with docetaxel and drugs acting on DNA in vitro, even after drug removal. Since cancer chemotherapy is typically administered during rest periods due to its high toxicity, combining it with an ALKBH3 inhibitor could be a promising strategy for enhancing cancer treatment, as it can elicit an additive effect during treatment, allowing dosage reduction, and synergistically sustain the effect after drug washout during rest periods.

3.
Nature ; 548(7666): 224-227, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28746308

ABSTRACT

Inhibitors of Mek1/2 and Gsk3ß, known as 2i, enhance the derivation of embryonic stem (ES) cells and promote ground-state pluripotency in rodents. Here we show that the derivation of female mouse ES cells in the presence of 2i and leukaemia inhibitory factor (2i/L ES cells) results in a widespread loss of DNA methylation, including a massive erasure of genomic imprints. Despite this global loss of DNA methylation, early-passage 2i/L ES cells efficiently differentiate into somatic cells, and this process requires genome-wide de novo DNA methylation. However, the majority of imprinting control regions (ICRs) remain unmethylated in 2i/L-ES-cell-derived differentiated cells. Consistently, 2i/L ES cells exhibit impaired autonomous embryonic and placental development by tetraploid embryo complementation or nuclear transplantation. We identified the derivation conditions of female ES cells that display 2i/L-ES-cell-like transcriptional signatures while preserving gamete-derived DNA methylation and autonomous developmental potential. Upon prolonged culture, however, female ES cells exhibited ICR demethylation regardless of culture conditions. Our results provide insights into the derivation of female ES cells reminiscent of the inner cell mass of preimplantation embryos.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Embryonic Stem Cells/cytology , Animals , Cell Differentiation/drug effects , DNA Methylation/drug effects , Embryonic Stem Cells/drug effects , Female , Genomic Imprinting/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Leukemia Inhibitory Factor/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Mice , Mice, Inbred C57BL
4.
Bioorg Med Chem Lett ; 30(1): 126744, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31759851

ABSTRACT

We carried out structure-activity relationship study on anti-cancer effects of naftopidil (1) and its metabolites, resulted in identification of 1-(4-hydroxy-2-methoxyphenyl)piperazin-1-yl)-3-(naphthalen-1-yloxy) propan-2-ol (2, HUHS190), a major human metabolite of 1, which exhibited the most selective toxicities between against normal and cancer cells (Table 1). 2 was more hydrophilic compared to 1, was enough to be prepared in high concentration solution of more than 100 µM in saline for an intravesical instillation drug. Moreover, serum concentration of 2 was comparable to that of 1, an oral preparation drug, after oral administration at 32 mg/kg (Fig. 3). Both of 1 and 2 showed broad-spectrum anti-cancer activities in vitro, for example, 1 and 2 showed inhibitory activity IC50 = 21.1 µM and 17.2 µM for DU145, human prostate cancer cells, respectively, and IC50 = 18.5 µM and 10.5 µM for T24 cells, human bladder cancer cells. In this study, we estimated anticancer effects of 2 in a bladder cancer model after intravesical administration similar to clinical cases. A single intravesical administration of 2 exhibited the most potent inhibitory activities among the clinical drugs for bladder cancers, BCG and Pirarubicin, without obvious side effects and toxicity (Fig. 4). Thus, HUHS190 (2) can be effective for patients after post-TURBT therapy of bladder cancer without side effects, unlike the currently available clinical drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Naphthalenes/therapeutic use , Piperazines/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Female , Humans , Male , Mice , Naphthalenes/pharmacology , Piperazines/pharmacology , Structure-Activity Relationship
5.
Proc Natl Acad Sci U S A ; 114(4): 758-763, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28057861

ABSTRACT

The spectrum of genetic mutations differs among cancers in different organs, implying a cellular context-dependent effect for genetic aberrations. However, the extent to which the cellular context affects the consequences of oncogenic mutations remains to be fully elucidated. We reprogrammed colon tumor cells in an ApcMin/+ (adenomatous polyposis coli) mouse model, in which the loss of the Apc gene plays a critical role in tumor development and subsequently, established reprogrammed tumor cells (RTCs) that exhibit pluripotent stem cell (PSC)-like signatures of gene expression. We show that the majority of the genes in RTCs that were affected by Apc mutations did not overlap with the genes affected in the intestine. RTCs lacked pluripotency but exhibited an increased expression of Cdx2 and a differentiation propensity that was biased toward the trophectoderm cell lineage. Genetic rescue of the mutated Apc allele conferred pluripotency on RTCs and enabled their differentiation into various cell types in vivo. The redisruption of Apc in RTC-derived differentiated cells resulted in neoplastic growth that was exclusive to the intestine, but the majority of the intestinal lesions remained as pretumoral microadenomas. These results highlight the significant influence of cellular context on gene regulation, cellular plasticity, and cellular behavior in response to the loss of the Apc function. Our results also imply that the transition from microadenomas to macroscopic tumors is reprogrammable, which underscores the importance of epigenetic regulation on tumor promotion.


Subject(s)
Adenomatous Polyposis Coli/genetics , Gene Expression Regulation/genetics , Genes, APC/physiology , Mutation/genetics , Alleles , Animals , Cell Lineage/genetics , Cell Plasticity/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Epigenesis, Genetic/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Intestinal Mucosa/metabolism , Mice , Pluripotent Stem Cells/metabolism
6.
Biochem Biophys Res Commun ; 519(4): 705-713, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31543342

ABSTRACT

ES cell (ESC) identity is stably maintained through the coordinated regulation of transcription factors and chromatin structure. SMARCB1, also known as INI1, SNF5, BAF47, is one of the subunits of SWI/SNF (BAF) complexes that play a crucial role in regulating gene expression by controlling chromatin dynamics. Genetic ablation of Smarcb1 in mice leads to embryonic lethality at the peri-implantation stage, indicating that Smarcb1 is important for the early developmental stages. However, the role of SMARCB1 in the maintenance of the ESC identity remains unknown. Here we established mouse ESCs lacking Smarcb1 and investigated the effect of Smarcb1 ablation on the differentiation propensity of ESCs. We found an increased expression of trophectoderm-related genes including Cdx2 in Smarcb1-deficient ESCs. Consistently, they exhibited an extended differentiation propensity into the trophectoderm lineage cells in teratomas. However, although Smarcb1-deficient cells were infrequently incorporated into the trophectoderm cell layer of blastocysts, they failed to contribute to mature placental tissues in vivo. Furthermore, Smarcb1-deficient cells exhibited a premature differentiation in the neural tissue of E14.5 chimeric embryos. Notably, we found that binding motifs for CTCF, which is involved in the maintenance of genomic DNA architecture was significantly enriched in chromatin regions whose accessibility was augmented in Smarcb1-deficient cells, while those for pluripotency factors were overrepresented in regions which have more closed structure in those cells. Collectively, we propose that SMARCB1-mediated remodeling of chromatin landscapes is important for the maintenance and differentiation of ESCs.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Mouse Embryonic Stem Cells/metabolism , SMARCB1 Protein/genetics , Animals , Cells, Cultured , Chromatin/metabolism , Embryonic Development/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mouse Embryonic Stem Cells/cytology , SMARCB1 Protein/metabolism
7.
Nat Chem Biol ; 13(6): 675-680, 2017 06.
Article in English | MEDLINE | ID: mdl-28437394

ABSTRACT

Target-protein degradation is an emerging field in drug discovery and development. In particular, the substrate-receptor proteins of the cullin-ubiquitin ligase system play a key role in selective protein degradation, which is an essential component of the anti-myeloma activity of immunomodulatory drugs (IMiDs), such as lenalidomide. Here, we demonstrate that a series of anticancer sulfonamides NSC 719239 (E7820), indisulam, and NSC 339004 (chloroquinoxaline sulfonamide, CQS) induce proteasomal degradation of the U2AF-related splicing factor coactivator of activating protein-1 and estrogen receptors (CAPERα) via CRL4DCAF15 mediated ubiquitination in human cancer cell lines. Both CRISPR-Cas9-based knockout of DCAF15 and a single amino acid substitution of CAPERα conferred resistance against sulfonamide-induced CAPERα degradation and cell-growth inhibition. Thus, these sulfonamides represent selective chemical probes for disrupting CAPERα function and designate DCAFs as promising drug targets for promoting selective protein degradation in cancer therapy.


Subject(s)
Indoles/pharmacology , Nuclear Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Sulfonamides/metabolism , Antineoplastic Agents/pharmacology , Gene Knockdown Techniques , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Proteolysis/drug effects , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Sulfonamides/pharmacology
8.
Bioorg Med Chem Lett ; 27(14): 3167-3172, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28576617

ABSTRACT

A series of 31 resveratrol derivatives was designed, synthesized and evaluated for activation and inhibition of the TRPA1 channel. Most acted as activators and desensitizers of TRPA1 channels like resveratrol or allyl isothiocyanate (AITC). Compound 4z (HUHS029) exhibited higher inhibitory activity than resveratrol with an IC50 value of 16.1µM. The activity of 4z on TRPA1 was confirmed in TRPA1-expressing HEK293 cells, as well as in rat dorsal root ganglia neurons by a whole cell patch clamp recording. Furthermore, pretreatment with 4z exhibited an analgesic effect on AITC-evoked TRPA1-related pain behavior in vivo.


Subject(s)
Analgesics/chemical synthesis , Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Stilbenes/chemistry , Transient Receptor Potential Channels/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Inhibitory Concentration 50 , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Pain/drug therapy , Patch-Clamp Techniques , Rats , Resveratrol , TRPA1 Cation Channel , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors
9.
Cell Physiol Biochem ; 37(1): 193-200, 2015.
Article in English | MEDLINE | ID: mdl-26289043

ABSTRACT

BACKGROUND/AIMS: The linoleic acid derivative DCP-LA selectively and directly activates PKCε. The present study aimed at understanding the mechanism of DCP-LA-induced PKCε activation. METHODS: Point mutation in the C2-like domain on PKCε was carried out, and each kinase activity was monitored in PC-12 cells using a föerster resonance energy transfer (FRET) probe with cyan fluorescent protein (CFP) and yellow fluorescent protein (YFP) at the N- and C-terminal ends of PKCε, respectively, or in the cell-free systems using a reversed phase high-performance liquid chromatography (HPLC). Intracellular PKCε mobilization was monitored in PC-12 cells using mRuby-conjugated PKCε. DCP-LA binding to PKCε was assayed using a fluorescein conjugated to DCP-LA at the carboxyl-terminal end (Fluo-DCP). Uptake of DCP-LA into cells was measured in PC-12 ells. RESULTS: In the FRET analysis, DCP-LA decreased the ratio of YFP signal intensity/CFP signal intensity in PC-12 cells and in the cell-free kinase assay, DCP-LA increased area of phosphorylated PKC substrate peptide, indicating DCP-LA-induced PKCε activation. These effects were significantly suppressed by replacing Arg50 and Ile89 by Ala or Asn in the C2-like domain of PKCε. In the fluorescent cytochemistry, DCP-LA did not affect intracellular PKCε distribution. In the cell-free binding assay, Fluo-DCP, that had no effect on the potential for PKCε activation, bound to PKCε, and the binding was inhibited only by mutating Ile89. Extracellularly applied DCP-LA was taken up into cells in a concentration-dependent manner. Although no activation was obtained in the cell-free kinase assay, the broad PKC activator PMA activated PKCε in PC-12 cells in association with translocation towards the cell surface, which was inhibited by mutating I89A. CONCLUSION: Unlike PMA DCP-LA activates cytosolic PKCε by binding to the phosphatidylserine binding/associating sites Arg50 and Ile89, possibly at the carboxyl-terminal end and the cyclopropane rings, respectively.


Subject(s)
Binding Sites/drug effects , Caprylates/pharmacology , Cytosol/drug effects , Phosphatidylserines/metabolism , Protein Kinase C/metabolism , Animals , Cell Line, Tumor , Cell-Free System/drug effects , Cell-Free System/metabolism , Cytosol/metabolism , Mutation/drug effects , PC12 Cells , Phosphorylation/drug effects , Protein Structure, Tertiary , Rats , Signal Transduction/drug effects
10.
Bioorg Med Chem Lett ; 25(16): 3373-7, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26099537

ABSTRACT

This Letter presents an effective method for the identification of target proteins of bioactive compounds such as drugs, natural products, and intrinsic ligands, using an affinity resin. The application of a photo-labile linker to an affinity resin enabled the selective elution of a target protein by irradiation for a short duration at 4 °C while leaving a large amount of non-specific binding protein on the resin. We have named this method the 'STEAP' method (selective target elution from affinity resins with photo-labile linker). Only a target protein that can bind the bioactive compound, the so-called 'active' protein, is eluted by the selective cleavage of the linker between the solid matrix and the target compound, and therefore, it is worth considering the potential of this method for the hyper-purification of proteins.


Subject(s)
Chemistry Techniques, Analytical/methods , Chromatography, Affinity , Drug Delivery Systems , Proteins/chemistry , Proteins/isolation & purification , Resins, Synthetic/chemistry , Photochemistry
11.
Bioorg Med Chem Lett ; 25(14): 2788-92, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26025877

ABSTRACT

Solid materials for affinity resins bearing long PEG spacers between a functional group used for immobilization of a bio-active compound and the solid surface were synthesized to capture not only small target proteins but also large and/or complex target proteins. Solid materials with PEG1000 or PEG2000 as spacers, which bear a benzenesulfonamide derivative, exhibited excellent selectivity between the specific binding protein carbonic anhydrase type II (CAII) and non-specific ones. These materials also exhibited efficacy in capturing a particular target at a maximal amount. Affinity resins using solid materials with PEG1000 or PEG2000 spacers, bear a FK506 derivative, successfully captured the whole target complex of specific binding proteins at the silver staining level, while all previously known affinity resins with solid materials failed to achieve this objective. These novel affinity resins captured other specific binding proteins such as dynamin and neurocalcin δ as well.


Subject(s)
Polyethylene Glycols/chemistry , Resins, Synthetic/chemistry , Tacrolimus/chemistry , Calcineurin/chemistry , Calcineurin/metabolism , Carbonic Anhydrase II/chemistry , Carbonic Anhydrase II/metabolism , Drug Design , Protein Binding , Sulfonamides/chemistry , Sulfonamides/metabolism , Tacrolimus/metabolism , Benzenesulfonamides
12.
J Pharmacol Sci ; 128(4): 179-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26238253

ABSTRACT

The phosphatidylethanolamine derivative 1,2-O-bis-[8-{2-(2-pentyl-cyclopropylmethyl)-cyclopropyl}-octanoyl]-sn-glycero-3-phosphatidylethanolamine (diDCP-LA-PE) promoted GLUT4 translocation to the cell surface in differentiated 3T3-L1-GLUT4myc adipocytes through a pathway along a phosphatidylinositol 3-kinase (PI3K)/3-phosphoinositide-dependent protein kinase-1 (PDK1)/Akt axis, that mimics insulin signaling. Moreover, diDCP-LA-PE-induced GLUT4 translocation was suppressed by inhibitors of the Rho GTPase Rac1 and Rho-associated coiled-coil-containing protein kinase (ROCK) or knocking-down Rac1 and ROCK1. The results of the present study show that Rac1 and ROCK are critical for regulation of GLUT4 trafficking by diDCP-LA-PE as well as insulin.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 4/metabolism , Phosphatidylethanolamines/pharmacology , Protein Transport/drug effects , Signal Transduction/physiology , rac1 GTP-Binding Protein/physiology , rho-Associated Kinases/physiology , 3-Phosphoinositide-Dependent Protein Kinases/physiology , Adipocytes/metabolism , Cell Line , Humans , Insulin , Phosphatidylinositol 3-Kinase/physiology , Proto-Oncogene Proteins c-akt/physiology
13.
Cancer Sci ; 105(7): 883-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24754309

ABSTRACT

The newly synthesized naftopidil analogue HUHS1015 reduced cell viability in malignant pleural mesothelioma cell lines MSTO-211H, NCI-H28, NCI-H2052, and NCI-H2452, with the potential greater than that for the anticancer drugs paclitaxel or cisplatin at concentrations higher than 30 µM. HUHS1015 induced both necrosis and apoptosis of MSTO-211H and NCI-H2052 cells. HUHS1015 upregulated expression of mRNAs for Puma, Hrk, and Noxa in MSTO-211H and NCI-H2052 cells, suggesting HUHS1015-induced mitochondrial apoptosis. HUHS1015 clearly suppressed tumor growth in mice inoculated with NCI-H2052 cells. Taken together, the results of the present study indicate that HUHS1015 could be developed as an effective anticancer drug for treatment of malignant pleural mesothelioma.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Pleural Neoplasms/drug therapy , Propanolamines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma, Malignant , Mice , Mice, Inbred BALB C , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation/drug effects
14.
Cell Physiol Biochem ; 33(2): 300-9, 2014.
Article in English | MEDLINE | ID: mdl-24525749

ABSTRACT

BACKGROUND/AIMS: The present study was conducted to understand biochemical and biological characteristics of the phosphatidylinositol (PI) derivative 1,2-O-bis-[8-{2-(2-pentyl-cyclopropylmethyl)-cyclopropyl}-octanoyl]-Sn-glycero-3-phosphatidyl-D-1-inositol (diDCP-LA-PI) and its enantiomer 1,2-O-bis-[8-{2-(2-pentyl-cyclopropylmethyl)-cyclopropyl}-octanoyl]-Sn-glycero-3-phosphatidyl-L-1-inositol (diDCP-LA-PIe), with 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA) on the α and ß position. METHODS: Activities of protein kinase C (PKC) and protein phosphatases such as protein phosphatase 1 (PP1), PP2A, and protein tyrosine phosphatase 1B (PTP1B) were assayed under the cell-free conditions and in PC-12 cells. Akt1/2 activity was monitored by quantifying phosphorylation at Thr308/309 and Ser473/474 in PC-12 cells. RESULTS: diDCP-LA-PI significantly activated PKCα, -ßΙ, -δ, and -ε, to an extent greater than that for diDCP-LA-PIe. diDCP-LA-PI still activated PKC in PC-12 cells, with the potential higher than that for diDCP-LA-PIe. Both diDCP-LA-PI and diDCP-LA-PIe reduced PP1 activity to a similar extent (30% of basal levels). diDCP-LA-PI enhanced PP2A activity to 180% of basal levels, while diDCP-LA-PIe had no effect. Drastic inhibition of PTP1B was obtained with diDCP-LA-PI and diDCP-LA-PIe, the extent reaching nearly 0% of basal levels. diDCP-LA-PI and diDCP-LA-PIe increased phosphorylation of Akt1/2 at Thr308/309 and Ser473/474 in PC-12 cells in the presence and absence of the PP2A inhibitor okadaic acid, respectively. CONCLUSION: The results of the present study show that diDCP-LA-PI and diDCP-LA-PIe exhibit different bioactivities with the different potentials each other.


Subject(s)
Phosphatidylinositols , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Animals , PC12 Cells , Phosphatidylinositols/chemical synthesis , Phosphatidylinositols/chemistry , Phosphatidylinositols/pharmacology , Phosphorylation/drug effects , Rats
15.
Cell Physiol Biochem ; 33(4): 1097-105, 2014.
Article in English | MEDLINE | ID: mdl-24732916

ABSTRACT

BACKGROUND/AIMS: The present study investigated the signaling pathway underlying Rac1 activation induced by the linoleic acid derivative 8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid (DCP-LA). METHODS: Activity of protein tyrosine phosphatase 1B (PTP1B) was assayed under cell-free conditions. Western blot was carried out to quantify phosphorylation of insulin receptor substrate-1 (IRS-1) and Akt in PC-12 cells. Rac1 activity was monitored in the föerster resonance energy transfer (FRET) analysis using living and fixed PC-12 cells. RESULTS: DCP-LA markedly suppressed PTP1B activity in a concentration (100 pM-100 µM)-dependent manner. In the DCP-LA binding assay, fluorescein-conjugated DCP-LA produced a single fluorescent signal band at 60 kDa, corresponding to the molecule of PTP1B, and the signal was attenuated or abolished by co-treatment or pretreatment with non-conjugated DCP-LA. DCP-LA significantly enhanced nerve growth factor (NGF)-stimulated phosphorylation of IRS-1 at Tyr1222 and Akt1/2 at Thr308/309 and Ser473/474 in PC-12 cells. In the FRET analysis, DCP-LA significantly enhanced NGF-stimulated Rac1 activation, which is abrogated by the phosphatidylinositol 3 kinase (PI3K) inhibitor wortmannin, the 3-phosphoinositide-dependent protein kinase-1 (PDK1) inhibitor BX912, or the Akt inhibitor MK2206. CONCLUSION: The results of the present study show that DCP-LA-induced PTP1B inhibition, possibly through its direct binding, causes Rac1 activation by enhancing a pathway along a receptor tyrosine kinase (RTK)/IRS-1/PI3K/Akt/Rac1 axis.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptor, trkA/metabolism , rac1 GTP-Binding Protein/metabolism , Androstadienes/pharmacology , Animals , Caprylates/chemistry , Caprylates/pharmacology , Fluorescein/chemistry , Fluorescence Resonance Energy Transfer , Heterocyclic Compounds, 3-Ring/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats , Signal Transduction/drug effects , Wortmannin
16.
Bioorg Med Chem Lett ; 24(4): 1071-4, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24461353

ABSTRACT

A series of 1-aryl-3,4-substituted-1H-pyrazol-5-ol derivatives was synthesized and evaluated as prostate cancer antigen-1 (PCA-1/ALKBH3) inhibitors to obtain a novel anti-prostate cancer drug. After modifying 1-(1H-benzimidazol-2-yl)-3,4-dimethyl-1H-pyrazol-5-ol (1), a hit compound found during random screening using a recombinant PCA-1/ALKBH3, 1-(1H-5-methylbenzimidazol-2-yl)-4-benzyl-3-methyl-1H-pyrazol-5-ol (35, HUHS015), was obtained as a potent PCA-1/ALKBH3 inhibitor both in vitro and in vivo. The bioavailability (BA) of 35 was 7.2% in rats after oral administration. As expected, continuously administering 35 significantly suppressed the growth of DU145 cells, which are human hormone-independent prostate cancer cells, in a mouse xenograft model without untoward effects.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Drug Design , Prostatic Neoplasms/drug therapy , Pyrazoles/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Prostatic Neoplasms/metabolism , Pyrazoles/administration & dosage , Pyrazoles/chemical synthesis , Rats , Structure-Activity Relationship
17.
Biochem J ; 450(2): 303-9, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23256752

ABSTRACT

In yeast two-hybrid screening, protein 4.1N, a scaffolding protein, was identified as a binding partner of the α7 ACh (acetylcholine) receptor. For rat hippocampal slices, the linoleic acid derivative DCP-LA {8-[2-(2-pentyl-cyclopropylmethyl)-cyclopropyl]-octanoic acid} increased the association of the α7 ACh receptor with 4.1N, and the effect was inhibited by GF109203X, an inhibitor of PKC (protein kinase C), although DCP-LA did not induce PKC phosphorylation of 4.1N. For PC-12 cells, the presence of the α7 ACh receptor in the plasma membrane fraction was significantly suppressed by knocking down 4.1N. DCP-LA increased the presence of the α7 ACh receptor in the plasma membrane fraction, and the effect was still inhibited by knocking down 4.1N. In the monitoring of α7 ACh receptor mobilization, DCP-LA enhanced signal intensities for the α7 ACh receptor at the membrane surface in PC-12 cells, which was clearly prevented by knocking down 4.1N. Taken together, the results of the present study show that 4.1N interacts with the α7 ACh receptor and participates in the receptor tethering to the plasma membrane. The results also indicate that DCP-LA increases membrane surface localization of the α7 ACh receptor in a 4.1N-dependent manner under the control of PKC, but without phosphorylating 4.1N.


Subject(s)
Caprylates/pharmacology , Cell Membrane/metabolism , Linoleic Acids/pharmacology , Receptors, Nicotinic/analysis , Receptors, Nicotinic/metabolism , Animals , HEK293 Cells , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Patch-Clamp Techniques , Rats , Rats, Wistar , Transfection , alpha7 Nicotinic Acetylcholine Receptor
18.
Molecules ; 19(12): 21462-72, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25532843

ABSTRACT

We have originally synthesized the naftopidil analogue 1-[2-(2-methoxyphenylamino)ethylamino]-3-(naphthalene-1-yloxy)propan-2-ol (HUHS 1015) as a new anticancer drug. HUHS1015 induces cell death in a wide variety of human cancer cell lines originated from malignant pleural mesothelioma, lung cancer, hepatoma, gastric cancer, colorectal cancer, bladder cancer, prostate cancer, and renal cancer. HUHS1015-induced cell death includes necrosis (necroptosis) and apoptosis, and the underlying mechanism differs depending upon cancer cell types. HUHS1015 effectively suppresses tumor growth in mice inoculated with NCI-H2052, MKN45, or CW2 cells, with a potential similar to or higher than that of currently used anticancer drugs. Here we show how HUHS1015 might offer brilliant hope for cancer therapy.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Propanolamines/pharmacology , Aniline Compounds/chemical synthesis , Aniline Compounds/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Humans , Mitochondrial Proteins/metabolism , Necrosis , Propanolamines/chemical synthesis , Propanolamines/therapeutic use , Xenograft Model Antitumor Assays
19.
J Med Ultrason (2001) ; 51(2): 347-353, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38581557

ABSTRACT

PURPOSE: The ratio of the internal carotid artery (ICA) to the common carotid artery (CCA), especially the "AcT ratio," which is a modified measurement method of acceleration time, is useful for diagnosing ICA-origin stenosis. However, previous studies were single-center studies. Therefore, this multicenter, retrospective, cross-sectional study aimed to determine whether a method using the AcT ratio is useful for estimating stenosis rates. METHODS: This study included 461 vessels subjected to carotid artery ultrasonography and evaluation for ICA-origin stenosis via NASCET at four hospitals. The duration from the steep rise point to the inflection point or the first peak was defined as AcT on pulsed wave Doppler. The AcT ratio was calculated as AcT of ICA/AcT of ipsilateral CCA. The AcT ratio and rate of ICA-origin stenosis were analyzed using Pearson's correlation coefficient, simple regression analysis, and ROC curve. RESULTS: A significant positive correlation was observed between the AcT ratio and NASCET stenosis. NASCET stenosis of ≥ 50% had a sensitivity, specificity, and negative predictive value (NPV) of 70.2%, 71.6%, and 91.5%, respectively, when the cut-off value of the AcT ratio was 1.17. NASCET stenosis of ≥ 70% had a sensitivity, specificity, and NPV of 70.5%, 72.1%, and 95.9%, respectively, when the cut-off value of the AcT ratio was 1.22. CONCLUSIONS: The findings of this multicenter, retrospective, cross-sectional study suggest that the AcT ratio is useful for diagnosing ICA-origin stenosis, especially for diagnosis by exclusion. NASCET stenosis of ≥ 50% was considered unlikely if the Act ratio was ≤ 1.17, whereas NASCET stenosis of ≥ 70% was considered unlikely if it was ≤ 1.22.


Subject(s)
Carotid Artery, Internal , Carotid Stenosis , Humans , Carotid Stenosis/diagnostic imaging , Retrospective Studies , Male , Cross-Sectional Studies , Female , Carotid Artery, Internal/diagnostic imaging , Aged , Middle Aged , Sensitivity and Specificity , Aged, 80 and over
20.
Cell Physiol Biochem ; 31(4-5): 555-64, 2013.
Article in English | MEDLINE | ID: mdl-23594444

ABSTRACT

BACKGROUND/AIMS: The linoleic acid derivative DCP-LA selectively activates PKCε and inhibits protein phosphatase 1 (PP1). In the present study, we have newly synthesized phosphatidyl-ethanolamine, -serine, -choline, and -inositol containing DCP-LA at the α and ß position (diDCP-LA-PE, -PS, PC, and -PI, respectively), and examined the effects of these compounds on activities of PKC isozymes and protein phosphatases. METHODS: Activities of PKC isozymes PKCα, -ßΙ, -ßΙΙ, -γ, -δ, -ε-, ι, and -ζ and protein phosphatases PP1, PP2A, and protein tyrosine phosphatase 1B (PTP1B) were assayed under the cell-free conditions. RESULTS: All the compounds activated PKC, with the different potential, but only PKCγ inhibition was obtained with diDCP-LA-PC. Of compounds diDCP-LA-PE alone significantly activated PKCι and -ζ. diDCP-LA-PE and diDCP-LA-PI suppressed PP1 activity, but otherwise diDCP-LA-PI enhanced PP2A activity. diDCP-LA-PE, diDCP-LA-PS, and diDCP-LA-PI strongly reduced PTP1B activity, while diDCP-LA-PC enhanced the activity. CONCLUSION: All the newly synthesized DCP-LA-phospholipids serve as a PKC activator and of them diDCP-LA-PE alone has the potential to activate the atypical PKC isozymes PKCι and -ζ. diDCP-LA-PE and diDCP-LA-PI serve as an inhibitor for PP1 and PTP1B, diDCP-LA-PS as a PTP1B inhibitor, diDCP-LA-PI as a PP2A enhancer, and diDCP-LA-PC as a PTP1B enhancer.


Subject(s)
Caprylates/chemistry , Enzyme Inhibitors/chemical synthesis , Phosphatidylethanolamines/chemistry , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Kinase C/chemistry , Caprylates/chemical synthesis , Caprylates/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Phosphatidylethanolamines/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Kinase C/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL