Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 15(12)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36558913

ABSTRACT

Metastasis, in which cancer cells migrate to other tissues and form new tumors, is a major cause of both cancer death and treatment failure. In a previous study, benproperine (Benp) was identified as a cancer cell migration inhibitor and an inhibitor of actin-related protein 2/3 complex subunit 2 (ARPC2). However, Benp is a racemic mixture, and which stereoisomer is the active isomer remains unclear. In this study, we found that S-Benp is an active isomer and inhibits the migration and invasion of cancer cells much more strongly than R-Benp, with no effect on normal cells. The metastasis inhibitory effect of S-Benp was also verified in an animal model. Validating that inhibitors bind to their targets in cells and tissues has been a very challenging task in drug discovery. The direct interactions between ARPC2 and S-Benp were verified by surface plasmon resonance analysis (SPR), a cellular thermal shift assay (CETSA), and drug affinity responsive target stability (DARTS). In the mutant study with ARPC2F225A cells, S-Benp did not bind to ARPC2F225A according to CETSA and DARTS. Furthermore, we validated that S-Benp colocalized with ARPC2 in cancer cells and directly bound to ARPC2 in tumor tissues using Cy3-conjugated S-Benp according to CETSA. Finally, actin polymerization assays and immunocytochemistry showed that S-Benp suppressed actin remodeling such as lamellipodium formation. Taken together, our data suggest that S-Benp is an active stereoisomer of Benp and a potential metastasis inhibitor via ARPC2 binding.

2.
Molecules ; 26(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34684783

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) plays a critical role in the formation and growth of human cancer. Therefore, STAT3 is a therapeutic target for cancer drug discovery. Acacetin, a flavone present in various plants, inhibits constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. Acacetin inhibits STAT3 activity by directly binding to STAT3, which we confirmed by a pull-down assay with a biotinylated compound and two level-free methods, namely, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). Acacetin inhibits STAT3 phosphorylation at the tyrosine 705 residue and nuclear translocation in DU145 cells, which leads to the downregulation of STAT3 target genes. Acacetin then induces apoptosis in a time-dependent manner. Interestingly, acacetin induces the production of reactive oxygen species (ROS) that are not involved in the acacetin-induced inhibition of STAT3 activation because the suppressed p-STAT3 level is not rescued by treatment with GSH or NAC, which are general ROS inhibitors. We also found that acacetin inhibits tumor growth in xenografted nude mice. These results suggest that acacetin, as a STAT3 inhibitor, could be a possible drug candidate for targeting STAT3 for the treatment of cancer in humans.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavones/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Protein Binding , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
3.
Cancers (Basel) ; 13(12)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203709

ABSTRACT

Although EGFR-TKI treatment of NSCLC (non-small-cell lung cancer) patients often achieves profound initial responses, the efficacy is transient due to acquired resistance. Multiple receptor tyrosine kinase (RTK) pathways contribute to the resistance of NSCLC to first- and third-generation EGFR-TKIs, such as erlotinib and osimertinib. To identify potential targets for overcoming EGFR-TKI resistance, we performed a gene expression signature-based strategy using connectivity map (CMap) analysis. We generated erlotinib-resistant HCC827-ErlR cells, which showed resistance to erlotinib, gefitinib, osimertinib, and doxorubicin. A list of differentially expressed genes (DEGs) in HCC827-ErlR cells was generated and queried using CMap analysis. Analysis of the top 4 compounds from the CMap list suggested HSF1 as a potential target to overcome EGFR-TKI resistance. HSF1 inhibition by using HSF1 shRNAs or KRIBB11 decreased the expression of HSF1 downstream proteins, such as HSP70 and HSP27, and also decreased the expression of HSP90/HSP70/BAG3 client proteins, such as BCL2, MCL1, EGFR, MET, and AXL, causing apoptosis of EGFR-TKI-resistant cancer cells. Finally, we demonstrated the efficacy of the HSF1 inhibitor on PC9-ErlR cells expressing mutant EGFR (T790M) in vivo. Collectively, these findings support a targetable HSF1-(HSP90/HSP70/BAG3)-(BCL2/MCL1/EGFR/MET/AXL) pathway to overcome multiple mechanisms of EGFR-TKI resistance.

4.
Phytother Res ; 35(3): 1508-1520, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33164240

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is aberrantly activated in many human cancers. We tried to find STAT3 inhibitors from natural sources and found that Xanthium fruit extracts decreased phosphorylation of STAT3-Y705. 8-Epi-xanthatin (EXT) was isolated from the extracts. When DU145 cancer cells were treated with EXT, p-STAT3-Y705 was decreased with an IC50 of 3.2 µM. EXT decreased the expression of STAT3 target genes, such as cyclin A, cyclin D1, and BCL-2, and induced PARP cleavage, indicating apoptotic cell death. Downregulation of EXT-induced p-STAT3-Y705 was rescued by pretreating DU145 cells with antioxidants, such as N-acetyl-L-cysteine (NAC), indicating that reactive oxygen species (ROS) were involved in the EXT-induced inhibition of STAT3 activation. Furthermore, we proved the association of EXT with STAT3 protein by using a drug affinity responsive target stability (DARTS) assay and a cellular thermal shift assay (CETSA). EXT inhibited proliferation of DU145 cells with a GI50 of 6 µM and reduced tumor growth in mice xenografted with DU145 cells. Immunoblotting showed that phosphorylation of STAT3-Y705 was lower in EXT-treated tumor tissue than in control tissues. Collectively, we found that EXT binds to, and inhibits, STAT3 activation and could be a lead compound for anticancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Fruit/chemistry , Furans/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , STAT3 Transcription Factor/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Furans/pharmacology , Humans , Male , Mice , Mice, Nude , Reactive Oxygen Species/metabolism , Signal Transduction
5.
Biochem Pharmacol ; 163: 46-59, 2019 05.
Article in English | MEDLINE | ID: mdl-30710516

ABSTRACT

Metastasis is the leading cause of cancer mortality and cancer cell migration is an essential stage of metastasis. We identified benproperine (Benp, a clinically used antitussive drug) as an inhibitor of cancer cell migration and an anti-metastatic agent. Benp selectively inhibited cancer cell migration and invasion, which also suppressed metastasis of cancer cells in animal models. Actin-related protein 2/3 complex subunit 2 (ARPC2) was identified as a molecular target of Benp by affinity column chromatography with Benp-tagged Sepharose beads. Benp bound directly to ARPC2 in cells, which was validated by pull-down assay using Benp-biotin and label-free biochemical methods such as the drug affinity responsive target stability (DARTS) and cellular thermal shift assay (CETSA). Benp inhibited Arp2/3 function, showing disruption of lamellipodial structure and inhibition of actin polymerization. Unlike Arp2/3 inhibitors, Benp selectively inhibited the migration of cancer cells but not normal cells. ARPC2-knockdown cancer cells showed defective cell migration and suppressed metastasis in an animal model. Therefore, ARPC2 is a potential target for anti-metastatic therapy, and Benp has the clinical potential to block metastasis. Furthermore, Benp is a useful agent for studying the functions of the Arp2/3 complex in cancer cell migration and metastasis.


Subject(s)
Actin-Related Protein 2-3 Complex/antagonists & inhibitors , Actin-Related Protein 2-3 Complex/metabolism , Antineoplastic Agents/pharmacology , Benzhydryl Compounds/pharmacology , Cell Movement/drug effects , Piperidines/pharmacology , Actin-Related Protein 2-3 Complex/chemistry , Animals , Antineoplastic Agents/therapeutic use , Benzhydryl Compounds/therapeutic use , Cell Movement/physiology , Dose-Response Relationship, Drug , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis/prevention & control , Piperidines/therapeutic use , Protein Structure, Secondary , Protein Structure, Tertiary , Xenograft Model Antitumor Assays/methods
6.
Cancer Sci ; 110(1): 366-378, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30375708

ABSTRACT

Inhibition of the signal transducer and activator of transcription 3 (STAT3) signaling pathway is a novel therapeutic strategy to treat human cancers with constitutively active STAT3. During the screening of natural products to find STAT3 inhibitors, we identified 2'-hydroxycinnamaldehyde (HCA) as a STAT3 inhibitor, which was isolated from the stem bark of Cinnamomum cassia. In this study, we found that HCA inhibited constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. HCA selectively inhibited the STAT3 activity by direct binding to STAT3, which was confirmed by biochemical methods, including a pull-down assay with biotin-conjugated HCA, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). HCA inhibited STAT3 phosphorylation at the tyrosine 705 residue, dimer formation, and nuclear translocation in DU145 cells, which led to a downregulation of STAT3 target genes. The downregulation of cell cycle progression and antiapoptosis-related gene expression by HCA induced the accumulation of cells in the G0/G1 phase of the cell cycle and then induced apoptosis. We also found that reactive oxygen species (ROS) were involved in the HCA-induced inhibition of STAT3 activation and cell proliferation because the suppressed p-STAT3 level was rescued by glutathione or N-acetyl-L-cysteine treatment, which are general ROS inhibitors. These results suggest that HCA could be a potent anticancer agent targeting STAT3-activated tumor cells.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Cinnamates/pharmacology , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cinnamates/chemistry , Female , HCT116 Cells , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA Interference , STAT3 Transcription Factor/genetics
7.
Cancer Lett ; 434: 42-55, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30009856

ABSTRACT

It is reported that 2'-hydroxycinnamaldehyde (HCA), isolated from cinnamon, has anti-tumor effects through the modulation of multi-target molecules. In this study, we identified pyruvate kinase M2 (PKM2) as a direct target of HCA by use of biochemical methods including affinity chromatography, drug affinity responsive target stability, and cellular thermal shift assay. PKM2 is up-regulated in multiple cancer types and is considered as a potential target for cancer therapy. HCA binds directly to PKM2 and selectively decreases the phosphorylation of PKM2 at Tyr105, indicating a potential anti-proliferative effect on prostate cancer cells. As a PKM2 activator, HCA increases pyruvate kinase activity by promoting the tetrameric state of PKM2. However, HCA suppresses protein kinase activity of PKM2 by decreasing the phosphorylation at Tyr105. Moreover, this leads to a decrease of PKM2-mediated STAT3 phosphorylation at Tyr705 and a down-regulation of target genes, including MEK5 and cyclin D1. Furthermore, HCA suppresses tumor growth and the release of tumor extracellular vesicles in vivo by inhibiting the phosphorylation of PKM2. Collectively, our results suggest that HCA may be a potential anticancer agent targeting PKM2 in cancer progression.


Subject(s)
Cell Proliferation/drug effects , Cinnamates/pharmacology , Prostatic Neoplasms/drug therapy , Pyruvate Kinase/antagonists & inhibitors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Animals , Cell Line , Cell Line, Tumor , HCT116 Cells , Humans , Male , Mice, Nude , PC-3 Cells , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Multimerization/drug effects , Pyruvate Kinase/chemistry , Pyruvate Kinase/metabolism , STAT3 Transcription Factor/metabolism
8.
Bioorg Med Chem Lett ; 28(14): 2566-2572, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29807795

ABSTRACT

Piperlongumine (PL), isolated from Piper longum L., is receiving intense interest due to its selectively ability to kill cancer cells but not normal cells. We synthesized a number of analogues by replacing the cyclic amide of PL with aliphatic amides to explore structural diversity. Compound CG-06 had the strongest cytotoxic profile of this series, showing potent effects in human prostate cancer DU-145 cells, in which signal transducer and activator of transcription 3 (STAT3) is constitutively active. CG-06 inhibited STAT3 phosphorylation at tyrosine 705 in a dose- and time dependent manner in DU-145 cells and suppressed IL-6-induced STAT3 phosphorylation at Tyr-705 in DU-145 and LNCaP cell lines. CG-06 decreased the expression levels of STAT3 target genes, such as cyclin A, Bcl-2, and survivin. Notably, we used drug affinity responsive target stability (DARTS) to show that CG-06 binds directly to STAT3, and the reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) rescued the CG-06-induced suppression p-STAT3. Our results suggest that CG-06 is a novel inhibitor of STAT3 and may be a useful lead molecule for the development of a therapeutic STAT3 inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Dioxolanes/pharmacology , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dioxolanes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Male , Molecular Structure , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship
9.
Carcinogenesis ; 39(6): 838-849, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29668859

ABSTRACT

We hypothesized that octamer-binding transcription factor 4 (OCT4) inhibition would have therapeutic benefits in testicular germ cell tumors (TGCT). To identify inhibitors of OCT4, a chemical library was screened using a luciferase reporter system under the control of an OCT4 response element. A compound named KRIBB53 was identified based on its blocking of OCT4-dependent luciferase activation. When NCCIT cells were exposed to KRIBB53, the expression levels of OCT4 target genes, such as NANOG and USP44, were inhibited with an IC50 of 13 and 15 µM, respectively. In addition, the levels of OCT4 were decreased by exposing NCCIT cells to KRIBB53, and pretreating the cells with the proteasomal inhibitor MG132 reversed the KRIBB53-induced OCT4 degradation. Biotinyl-KRIBB53 was synthesized and showed comparable activity to KRIBB53 in OCT4 downregulation. Using affinity chromatography assay, KRIBB53 was shown to associate with OCT4 in vitro. Furthermore, the drug affinity responsive target stability (DARTS) assay confirmed unmodified KRIBB53 binding to OCT4. KRIBB53 selectively inhibited proliferation of TGCT cells such as NCCIT and Tera-1 cells but not that of immortalized normal cells. Finally, the administration of KRIBB53 at 30 mg/kg reduced tumor volumes by 77% in the mice xenografted with NCCIT cells relative to their vehicle-treated counterparts. Immunoblotting assays showed that expression of OCT4 was lower in KRIBB53-treated tumor tissues than in control tissues. We provide the first report, to our knowledge, of an OCT4 inhibitor that binds to OCT4 and induces its degradation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms, Germ Cell and Embryonal/drug therapy , Octamer Transcription Factor-3/metabolism , Proteasome Endopeptidase Complex/metabolism , Testicular Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Mice, Inbred NOD , Neoplasms, Germ Cell and Embryonal/metabolism , Response Elements/drug effects , Testicular Neoplasms/metabolism , Ubiquitin-Specific Proteases/metabolism
10.
Bioorg Med Chem Lett ; 28(5): 853-857, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29456111

ABSTRACT

During the search for signal transducer and activator of transcription 3 (STAT3) inhibitors from natural products, methyllucidone, isolated from Lindera species (Lauraceae), was identified as a STAT3 inhibitor. Methyllucidone inhibited STAT3 phosphorylation at tyrosine 705 in a dose- and time dependent manner in DU145 prostate cancer cells and suppressed IL-6-induced STAT3 phosphorylation at Tyr-705 in LNCaP cells. Methyllucidone decreased the expression levels of STAT3 target genes, such as cyclin D1, cyclin A, Bcl-2, Mcl-1, and survivin. Methyllucidone inhibited DU145 cell growth and induced apoptosis by arresting the cell cycle at G1 phase. Notably, knockdown of the MEG2 gene by small interfering RNA suppressed the ability of methyllucidone to inhibit STAT3 activation. Methyllucidone regulates STAT3 activity by modulating MEG2 expression, and our results suggest that this compound is a novel inhibitor of the STAT3 pathway and may be a useful lead molecule for the development of a therapeutic STAT3 inhibitor.


Subject(s)
Cyclopentanes/pharmacology , Prostatic Neoplasms/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Cyclopentanes/chemistry , Cyclopentanes/isolation & purification , Dose-Response Relationship, Drug , Humans , Lauraceae/chemistry , Male , Molecular Structure , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Int J Oncol ; 51(4): 1331-1342, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28902363

ABSTRACT

Ginkgetin has been reported to display antitumor activity. However, the relevant pathway integrating cell cycle regulation and signaling pathways involved in growth inhibition in CRC cells remains to be identified. In this study, ginkgetin-treated HCT116 CRC cells exhibited significant dose-dependent growth inhibition with a GI50 value of 4.0 µM for 48-h treatment, together with apoptosis, via G2-phase cell cycle arrest. When HCT116 cells were treated with 10 µM ginkgetin for 48 h, the percentage of cells in G2/M phase increased by 2.2-fold (43.25%) versus the untreated control (19.69%). Ginkgetin regulated the expression of genes that are critically involved in G2 phase arrest cells, such as b­Myb, CDC2 and cyclin B1. Furthermore, we found that the suppression of b­Myb expression by ginkgetin was rescued ~5.1-fold by treatment with a miR-34a inhibitor (500 nM) and b­Myb was downregulated by >80% by 100 nM miR­34a mimic. These data suggest that the miRNA34a/b­Myb/cyclin B1 cascade plays a critical role in ginkgetin-induced G2 cell cycle arrest, as well as in the inhibition of HCT116 cell proliferation. Moreover, the administration of ginkgetin (10 mg/kg) reduced tumor volumes by 36.5% and tumor weight by 37.6% in the mice xenografted with HCT116 cells relative to their vehicle-treated counterparts. Therefore, ginkgetin is the first compound shown to regulate b­Myb by modulating miR-34a, and we suggest the use of ginkgetin as an inducer of G2 arrest for the treatment of CRC.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Biflavonoids/administration & dosage , Cell Cycle Proteins/genetics , Colonic Neoplasms/drug therapy , MicroRNAs/genetics , Trans-Activators/genetics , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Biflavonoids/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Xenograft Model Antitumor Assays
12.
Biochem Pharmacol ; 142: 46-57, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28666623

ABSTRACT

The roles and significance of signal transducer and activator of transcription 3 (STAT3) in human cancers have been extensively studied and STAT3 is a promising therapeutic target for cancer drug discovery. During the screening of natural products to identify STAT3 inhibitors, we identified geranylnaringenin (CG902), which decreased luciferase activity in a dose-dependent manner. CG902 specifically inhibited STAT3 phosphorylation at Tyr-705 in DU145 prostate cancer cells and decreased the expression levels of STAT3 target genes, such as cyclin D1, cyclin A, and survivin. Notably, the knockdown of the SHP-2 gene by small interfering RNA suppressed the ability of CG902 to inhibit STAT3 activation and CG902 activated the phosphatase activity of SHP-2 through direct interaction with SHP-2 and induced the phosphorylation of SHP-2. The interactions between CG902 and SHP-2 were confirmed by pull-down assay using biotinylated CG902. The interactions were also further validated by the drug affinity responsive target stability (DARTS) and cellular thermal shift assay (CETSA). The inhibitory effect of CG902 on cell growth was confirmed using the DU145 mouse xenograft model. We propose that CG902 inhibits STAT3 activity through a mechanism that involves the interactions between CG902 and SHP-2, and the phosphorylation of SHP-2, which leads to SHP-2 activation in DU145 cells. CG902 is the first compound to regulate STAT3 activity via the modulation of SHP-2 activity, and our results suggest that CG902 is a novel inhibitor of the STAT3 pathway and an activator of SHP-2, and may be a useful lead molecule for the development of a therapeutic STAT3 inhibitor.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Flavanones/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Artocarpus/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Flavanones/therapeutic use , Flow Cytometry , Humans , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Plant Leaves/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Time Factors , Transfection , Xenograft Model Antitumor Assays
13.
Int J Biochem Cell Biol ; 81(Pt A): 57-66, 2016 12.
Article in English | MEDLINE | ID: mdl-27780754

ABSTRACT

PPARγ agonists induced obesity in animal models as a side effect. Microarray experiments reveal that PPARγ agonist upregulates the expression of lipin-1 and this upregulation is correlated with the activity of the agonists. Lipin-1 induced by PPARγ agonists decreased the levels of PPARγ and ERK1/2 phosphorylation through direct interaction with these proteins in 3T3-L1 cells. In PPARγ agonist-treated 3T3-L1 preadipocytes, the knockdown of lipin-1 expression by small interfering RNA inhibited the adipogenesis that was induced by PPARγ agonists. In contrast, PPARγ2 expression was increased, and lipid droplets were accumulated in lipin-1-overexpressing 3T3-L1 adipocytes. Rosiglitazone (RGZ), a strong PPARγ agonist, synergistically promoted PPARγ dephosphorylation and adipogenesis in lipin-1-overexpressing 3T3-L1 preadipocytes. Therefore, lipin-1 has dual functions as a transcriptional cofactor and phosphatidate phosphatase (PAP) in the differentiation of preadipocyte cells induced by strong PPARγ agonists. In addition, the adipogenesis of 3T3-L1 cells was markedly upregulated by diacylglycerol (DAG), which was produced by lipin-1. Therefore, lipin-1 induction by PPARγ agonists might be an important factor in understanding the biological mechanism of the agonists' adverse effects, and this information may be valuable in the development of type-2 diabetes mellitus (T2DM) therapeutics with reduced adverse effects and greater tolerability.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Nuclear Proteins/genetics , PPAR gamma/agonists , Phosphatidate Phosphatase/genetics , Thiazolidinediones/pharmacology , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Diglycerides/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Rosiglitazone
14.
Phytother Res ; 30(5): 754-67, 2016 May.
Article in English | MEDLINE | ID: mdl-26890810

ABSTRACT

Cinnamaldehyde and cinnamaldehyde-derived compounds are candidates for the development of anticancer drugs that have received extensive research attention. In this review, we summarize recent findings detailing the positive and negative aspects of cinnamaldehyde and its derivatives as potential anticancer drug candidates. Furthermore, we describe the in vivo pharmacokinetics and metabolism of cinnamaldehydes. The oxidative and antioxidative properties of cinnamaldehydes, which contribute to their potential in chemotherapy, have also been discussed. Moreover, the mechanism(s) by which cinnamaldehydes induce apoptosis in cancer cells have been explored. In addition, evidence of the regulatory effects of cinnamaldehydes on cancer cell invasion and metastasis has been described. Finally, the application of cinnamaldehydes in treating various types of cancer, including breast, prostate, and colon cancers, has been discussed in detail. The effects of cinnamaldehydes on leukemia, hepatocellular carcinoma, and oral cancer have been summarized briefly. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Acrolein/analogs & derivatives , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Acrolein/administration & dosage , Acrolein/therapeutic use , Humans
15.
Biochem Pharmacol ; 97(1): 38-50, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26212545

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in various human cancers and has been used as a therapeutic target for tumors. This study screened natural products to identify compounds that inhibit STAT3 activity using a STAT3-dependent luciferase reporter system. Sugiol was identified as a compound that decreased luciferase activity in a dose-dependent manner. Sugiol specifically inhibited STAT3 phosphorylation at Tyr-705 in DU145 prostate cells, and this inhibition was independent of the STAT3 upstream kinase. Sugiol induced cell cycle arrest and decreased the expression levels of STAT3 target genes, such as cyclin D1, cyclin A, and survivin. Notably, we observed that sugiol interacted with transketolase, an enzyme in central metabolism, and increased ROS levels leading to the activation of ERK, which inhibits STAT3 activity. The protein phosphatase MEG2 was also responsible for sugiol-induced STAT3 dephosphorylation. The inhibitory effect of sugiol on cell growth was confirmed using the DU145 mouse xenograft model. We propose that sugiol inhibits STAT3 activity through a mechanism that involves the inhibition of transketolase, which leads to increased ROS levels and MEG2 activation in DU145 cells. Therefore, sugiol is the first compound regulating STAT3 activity via modulation of cancer metabolic pathway and we suggest the use of sugiol as an inhibitor of the STAT3 pathway for the treatment of human solid tumors with activated STAT3.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , Enzyme Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Reactive Oxygen Species/agonists , STAT3 Transcription Factor/antagonists & inhibitors , Transketolase/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma/drug therapy , Carcinoma/enzymology , Carcinoma/metabolism , Cell Line, Tumor , Diterpenes/therapeutic use , Enzyme Activation/drug effects , Enzyme Inhibitors/therapeutic use , Female , Genes, Reporter/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/chemistry , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/agonists , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Transketolase/chemistry , Transketolase/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Carcinogenesis ; 36(6): 696-706, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25840992

ABSTRACT

Heat shock factor 1 (HSF1) is a transcription factor for heat shock proteins (HSPs) expression that enhances the survival of cancer cells exposed to various stresses. HSF1 knockout suppresses carcinogen-induced cancer induction in mice. Therefore, HSF1 is a promising therapeutic and chemopreventive target. We performed cell-based screening with a natural compound collection and identified fisetin, a dietary flavonoid, as a HSF1 inhibitor. Fisetin abolished heat shock-induced luciferase activity with an IC50 of 14 µM in HCT-116 cancer cells. The treatment of HCT-116 with fisetin inhibited proliferation with a GI50 of 23 µM. When the cells were exposed to heat shock in the presence of fisetin, the induction of HSF1 target proteins, such as HSP70, HSP27 and BAG3 (Bcl-2-associated athanogene domain 3), were inhibited. HSP70/BAG3 complexes protect cancer cells from apoptosis by stabilizing anti-apoptotic Bcl-2 family proteins. The downregulation of HSP70/BAG3 by fisetin significantly reduced the amounts of Bcl-2, Bcl-xL and Mcl-1 proteins, subsequently inducing apoptotic cell death. Chromatin immunoprecipitation assays showed that fisetin inhibited HSF1 activity by blocking the binding of HSF1 to the hsp70 promoter. Intraperitoneal treatment of nude mice with fisetin at 30mg/kg resulted in a 35.7% (P < 0.001) inhibition of tumor growth.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Flavonoids/pharmacology , Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/biosynthesis , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Flavonols , HCT116 Cells , HSP27 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Proteins , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Chaperones , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Neoplasms/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , bcl-X Protein/biosynthesis
17.
Phytother Res ; 29(5): 749-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25682949

ABSTRACT

Artocarpus altilis (Parkinson) Fosberg has traditionally been used in Indonesia for the treatment of liver cirrhosis, hypertension, and diabetes. In many other countries, it is used for the treatment of malaria, yellow fever, and dengue fever. It has been reported that A. altilis extracts have antiatherosclerotic and cytoprotective effects, but its molecular targets in tumor cells are not yet fully understood. The A. altilis extracts and the partially purified fraction have been shown to inhibit STAT3 activity and the phosphorylation of STAT3 in a dose-dependent manner. To identify the active components, a bioassay-guided isolation of the partially purified fraction resulted in the identification of a geranyl dihydrochalcone, CG901. Its chemical structure was established on the basis of spectroscopic evidence and comparison with published data. The partially purified fraction and the isolated a geranyl dihydrochalcone, CG901, down-regulated the expression of STAT3 target genes, induced apoptosis in DU145 prostate cancer cells via caspase-3 and PARP degradation, and inhibited tumor growth in human prostate tumor (DU145) xenograft initiation model. These results suggest that A. altilis could be a good natural source and that the isolated compound will be a potential lead molecule for developing novel therapeutics against STAT3-related diseases, including cancer and inflammation.


Subject(s)
Artocarpus/chemistry , Chalcones/pharmacology , Plant Extracts/pharmacology , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor/drug effects , Female , Humans , Male , Mice, Inbred BALB C , Phosphorylation , Plant Leaves/chemistry , Xenograft Model Antitumor Assays
18.
Cancer Sci ; 106(4): 413-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25611086

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in human cancers. Therefore, STAT3 is a therapeutic target of cancer drug discovery. We previously reported that natural products inhibited constitutively activated STAT3 in human prostate tumor cells. We used a dual-luciferase assay to screen 200 natural products isolated from herbal medicines and we identified ginkgetin obtained from the leaves of Ginkgo biloba L. as a STAT3 inhibitor. Ginkgetin inhibited both inducible and constitutively activated STAT3 and blocked the nuclear translocation of p-STAT3 in DU-145 prostate cancer cells. Furthermore, ginkgetin selectively inhibited the growth of prostate tumor cells stimulated with activated STAT3. Ginkgetin induced STAT3 dephosphorylation at Try705 and inhibited its localization to the nucleus, leading to the inhibition of expression of STAT3 target genes such as cell survival-related genes (cyclin D1 and survivin) and anti-apoptotic proteins (Bcl-2 and Bcl-xL). Therefore, ginkgetin inhibited the growth of STAT3-activated tumor cells. We also found that ginkgetin inhibited tumor growth in xenografted nude mice and downregulated p-STAT3(Tyr705) and survivin in tumor tissues. This is the first report that ginkgetin exerts antitumor activity by inhibiting STAT3. Therefore, ginkgetin is a good STAT3 inhibitor and may be a useful lead molecule for development of a therapeutic STAT3 inhibitor.


Subject(s)
Antineoplastic Agents/therapeutic use , Biflavonoids/therapeutic use , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/biosynthesis , Drug Screening Assays, Antitumor , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Ginkgo biloba/metabolism , HCT116 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Prostate/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Repressor Proteins/biosynthesis , STAT3 Transcription Factor/biosynthesis , Survivin , Xenograft Model Antitumor Assays
19.
Biochem Pharmacol ; 89(1): 62-73, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24607275

ABSTRACT

The constitutive activation of STAT3 in human cancers causes the abnormal proliferation and survival of cancer cells, and thus, STAT3 is a therapeutic target of antitumor drugs. We screened a small-molecule library of 8600 synthetic compounds from the "Korea Chemical Bank" to identify inhibit STAT3 activity using a cell-based luciferase assay system. KT-18618 ((Z)-N-(4-chlorophenyl)-N-methyl-2-[1,3,3,3,-tetrafluoro-2-(thiophen-2-yl)prop-1-enyloxy]-acetamide) was selected as a novel inhibitor of the JAK/STAT3 pathway. KT-18618 inhibited STAT3 phosphorylation and the expression of STAT3-regulated genes. The inhibition of STAT3 phosphorylation led to the apoptosis of MDA-MB-468 cells. We postulated that the inhibition of the JAK family of proteins or c-Src inhibited STAT3 phosphorylation. Interestingly, the phosphorylation of these kinases was only mildly inhibited, but the phosphorylation of STAT3 was completely inhibited. This result implies that the inhibition of STAT3 phosphorylation by KT-18618 is an independent event that occurs through the phosphorylation of upstream kinases. Co-immunoprecipitation experiments revealed that KT-18618 inhibited the JAK3-STAT3 interaction. Moreover, JAK3 molecules were captured by biotinylated KT-18618, implying that KT-18618 bound to JAK3 molecules. Additionally, 1µM KT-18618 inhibited JAK3 kinase activity by approximately 28% in an in vitro kinase assay. From these results, we suggest that KT-18618 binds to JAK3 molecules and disrupts the JAK3-STAT3 interaction, which leads to the inhibition of STAT3 phosphorylation. KT-18618 is the first inhibitor of the JAK3-STAT3 interaction.


Subject(s)
Janus Kinase 3/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Flow Cytometry , Heterografts , Humans , Mice , Mice, Nude , Phosphorylation , Protein Binding , STAT3 Transcription Factor/metabolism
20.
J Biol Chem ; 288(40): 28713-26, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23983126

ABSTRACT

Heat shock factor 1 (HSF1) enhances the survival of cancer cells under various stresses. The knock-out of HSF1 impairs cancer formation and progression, suggesting that HSF1 is a promising therapeutic target. To identify inhibitors of HSF1 activity, we performed cell-based screening with a library of marketed and experimental drugs and identified cantharidin as an HSF1 inhibitor. Cantharidin is a potent antitumor agent from traditional Chinese medicine. Cantharidin inhibited heat shock-induced luciferase activity with an IC50 of 4.2 µm. In contrast, cantharidin did not inhibit NF-κB luciferase reporter activity, demonstrating that cantharidin is not a general transcription inhibitor. When the HCT-116 colorectal cancer cells were exposed to heat shock in the presence of cantharidin, the induction of HSF1 downstream target proteins, such as HSP70 and BAG3 (Bcl-2-associated athanogene domain 3), was suppressed. HSP70 and its co-chaperone BAG3 have been reported to protect cells from apoptosis by stabilizing anti-apoptotic Bcl-2 family proteins. As expected, treating HCT-116 cancer cells with cantharidin significantly decreased the amounts of BCL-2, BCL-xL, and MCL-1 protein and induced apoptotic cell death. Chromatin immunoprecipitation analysis showed that cantharidin inhibited the binding of HSF1 to the HSP70 promoter and subsequently blocked HSF1-dependent p-TEFb recruitment. Therefore, the p-TEFb-dependent phosphorylation of the C-terminal domain of RNA polymerase II was blocked, arresting transcription at the elongation step. Protein phosphatase 2A inhibition with PP2CA siRNA or okadaic acid did not block HSF1 activity, suggesting that cantharidin inhibits HSF1 in a protein phosphatase 2A-independent manner. We show for the first time that cantharidin inhibits HSF1 transcriptional activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cantharidin/pharmacology , DNA-Binding Proteins/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/pathology , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Acetylation/drug effects , Apoptosis Regulatory Proteins , Cantharidin/chemistry , Cantharidin/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/drug effects , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Humans , Mitosis/drug effects , Mitosis/genetics , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Positive Transcriptional Elongation Factor B , Protease Inhibitors/pharmacology , Protein Binding/drug effects , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...