Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 24(11)2023 May 27.
Article in English | MEDLINE | ID: mdl-37298331

ABSTRACT

Proteasome inhibitors (PIs) have emerged as an attractive novel cancer therapy. However, most solid cancers are seemingly resistant to PIs. The activation of transcription factor Nuclear factor erythroid 2 related factor-1 (NFE2L1) has been characterized as a potential resistance response to protect and restore proteasome activity in cancer cells. In this study, we demonstrated that α-Tocotrienol (T3) and redox-silent analogs of vitamin E (TOS, T3E) enhanced the sensitivity of bortezomib (BTZ), a proteasome inhibitor, in solid cancers through modulation of NFE2L1. In BTZ treatment, all of T3, TOS, and T3E inhibited an increase in the protein levels of NFE2L1, the expression levels of proteasome-related proteins, as well as the recovery of proteasome activity. Moreover, the combination of one of T3, TOS, or T3E and BTZ induced a significant decrease in cell viability in solid cancer cell lines. These findings suggested that the inactivation of NFE2L1 by T3, TOS, and T3E is essential to potentiate the cytotoxic effect of the proteasome inhibitor, BTZ, in solid cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Bortezomib/pharmacology , Proteasome Inhibitors/pharmacology , Vitamin E/pharmacology , Proteasome Endopeptidase Complex/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Oxidation-Reduction , Drug Resistance, Neoplasm , Neoplasms/drug therapy
2.
Biosci Biotechnol Biochem ; 84(3): 526-535, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31743080

ABSTRACT

Lysyl oxidase (LOX) is required for the formation of bone collagen cross-links. Inactivation of the LOX gene in osteoblasts by DNA methylation and JAK signaling has been reported to cause loss of cross-links and an increased risk of fractures. Tocotrienols (T3s) have proven benefits on bone strength, but their potential effects on LOX remain largely unknown. Thus, the present study investigates the in vitro effects of T3s on LOX expression in human osteoblastic MG-63 cells. Results indicated that Tocotrienol-Rich Fraction (TRF), the δ-T3 rich oil extracted from Annatto was the most effective and significantly increased LOX expression. TRF treatment decreased de-novo methyltransferases (DNMTs), DNMT3A and DNMT3B levels. In addition, TRF significantly inhibited JAK2 activation and decreased expression of Fli1, a transcription factor of DNMTs. We conclude that TRF induced an increase in LOX expression via inhibition of de-novo methylation and reduction of Fli1 expression by the inactivation of JAK2.Abbreviations: CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; Fli1: friend leukemia virus integration 1; JAK: janus kinase; LOX: lysyl oxidase; PCR: polymerase chain reaction; STAT: signal transducers and activators of transcription; T3s: tocotrienols; TPs: tocopherols; TRF: Tocotrienol-Rich Fraction.


Subject(s)
Bixaceae/metabolism , Carotenoids/metabolism , Osteoblasts/metabolism , Plant Extracts/metabolism , Protein-Lysine 6-Oxidase/metabolism , Tocotrienols/metabolism , Cell Line , Humans , Osteoblasts/enzymology
3.
Pharmacology ; 102(1-2): 26-36, 2018.
Article in English | MEDLINE | ID: mdl-29763912

ABSTRACT

BACKGROUND: Wnt signaling plays an essential role in tumor cell growth, including the development of malignant mesothelioma (MM). Epigenetic silencing of negative Wnt regulators leading to constitutive Wnt signaling has been observed in various cancers and warrants further attention. We have reported that a succinate ether derivative of α-tocotrienol (T3E) has potent cytotoxic effects in MM cells. Thus, in this study, we investigated whether the anti-MM effect of T3E could be mediated via the epigenetic alteration of the Wnt antagonist gene, Dickkopf-1 (DKK1). METHODS: WST-1 and cell analyzers were employed to analyze the effects of T3E on cell viability and apoptosis of human MM cell lines (H2452, H28). Real-time PCR and Western blot were performed to evaluate the expression at mRNA and protein levels. Methylation status and epigenetic modifications of DKK1's promoter regions after T3E treatment in MM cells were studied using methylation-specific PCR and Chromatin immunoprecipitation. Small interfering RNA-mediated knockdown -(siRNA), and specific inhibitors, were used to validate DKK1 as a target of T3E. RESULTS: T3E markedly impaired MM cell viability, increased the expression of phosphorylated-JNK and DKK1 and suppressed cyclin D, a downstream target gene of Wnt signaling. Knockdown of DKK1 expression by siRNA or a specific JNK inhibitor confirmed the contribution of DKK1 and JNK to T3E-induced cytotoxicity in MM cells. On the other hand, cytoskeleton-associated protein 4 (CKAP4) expression, which promotes cell proliferation as a Wnt-independent DKK1 receptor was inhibited by T3E. Silencing CKAP4 by -siRNA did not appear to directly affect MM cell viability, thereby indicating that expression of both DKK1 and CKAP4 is required. Furthermore, T3E-mediated inhibition of both DNA methyltransferases (DNMT1, 3A, and 3B) and histone deacetylases (HDAC1, 2, 3, and 8) in MM cells leads to increased DKK1 expression, thereby promoting tumor growth inhibition. MM cells treated with Zebularine (a DNMT inhibitor) and sodium butyrate (an HDAC inhibitor) exhibited cytotoxic effects, which may explain the inhibitory action of T3E on MM cells. In addition, an enhanced expression of DKK1 in MM cells following T3E treatment is positively correlated with the methylation status of its promoter; T3E decreased DNA methylation and increased histone acetylation. Moreover, T3E specifically increased histone H3 lysine 4 (H3K4) methylation activity, whereas no effects were observed on histone H3K9 and H3K27. CONCLUSIONS: Targeting the epigenetic induction of DKK1 may lead to effective treatment of MM, and T3E has great potential to induce anti-MM activity.


Subject(s)
Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Intercellular Signaling Peptides and Proteins/biosynthesis , Lung Neoplasms/genetics , Mesothelioma/genetics , Tocotrienols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/biosynthesis , Mesothelioma, Malignant , RNA, Small Interfering/pharmacology
4.
Anticancer Drugs ; 28(3): 289-297, 2017 03.
Article in English | MEDLINE | ID: mdl-27926612

ABSTRACT

Prostate cancer (PCa) is one of the most common cancers in Western countries and acquires a malignant phenotype, androgen-independent growth. PCa under hypoxia often has resistance to chemotherapy and radiotherapy. However, an effective therapy against PCa under hypoxia has not yet been established. In this report, we investigated the inhibitory effect of a redox-silent analogue of tocotrienol on the survival of a human androgen-independent PCa cell line (PC3) under hypoxia. We found that the redox-silent analogue exerted a cytotoxic effect on PC3 cells in a dose-dependent manner irrespective of either hypoxia or normoxia. Moreover, under hypoxia, the analogue dose dependently reduced the protein levels of hypoxia-inducible factor (HIF)-1α and HIF-2α. In addition, a specific inhibitor toward HIF-1α induced cytotoxicity on PC3 cells, whereas selective inhibition of HIF-2α exerted no effect. Furthermore, suppression of HIFs levels by the analogue in hypoxic PC3 cells was closely associated with the inactivation of Fyn, a member of the nonreceptor tyrosine kinase family, as confirmed by the action of a specific inhibitor toward the kinase (PP2). Taken together, these results suggest that the tocotrienol analogue could inhibit the survival of PC3 cells under hypoxia, mainly by the inhibition of Fyn/HIF-1α signaling, and this may lead to the establishment of a new effective therapy for androgen-independent PCa.


Subject(s)
Cell Hypoxia/drug effects , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Tocotrienols/pharmacology , Adaptation, Physiological/drug effects , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Oxidation-Reduction , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/metabolism
5.
Biol Pharm Bull ; 37(5): 865-70, 2014.
Article in English | MEDLINE | ID: mdl-24790010

ABSTRACT

Vascular endothelial growth factor (VEGF) plays a crucial role in tumor angiogenesis and represents an attractive anticancer target. We have previously demonstrated that a redox-silent analogue of α-tocotrienol, 6-O-carboxypropyl-α-tocotrienol (T3E) exhibits potent anti-carcinogenic property in human malignant mesothelioma (MM) cells. However, inhibition of tumor growth by targeting VEGF pathway remains undetermined. In this study, we explored the inhibitory effect of T3E on the paracrine secretion of VEGF in MM cells under mimicked hypoxia by cobalt chloride (CoCl2). In this study we examine whether T3E can suppress the secretion of VEGF in MM cells exposed to mimic hypoxia by cobalt chloride (CoCl2). We found that CoCl2-induced hypoxia treatment leads to increased up-regulated hypoxia-inducible factor-2α (HIF-2α) and subsequently induced the secretion of VEGF in MM cells. This up-regulation activation mainly depended on the activation of Yes, a member of the Src family of kinases. Treatment of hypoxic MM cells with T3E effectively inhibited the secretion of VEGF, On the other hand, T3E inhibited CoCl2-induced gene expression of VEGF due to the inactivation of Yes/HIF-2α signaling. These data suggest that Yes/HIF2-α/VEGF could be a promising therapeutic target of T3E in MM cells.


Subject(s)
Cobalt/pharmacology , Mesothelioma/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Signal Transduction/drug effects , Tocotrienols/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression/drug effects , Humans , Hypoxia/chemically induced , Hypoxia/metabolism , Mesothelioma/pathology , Oxidation-Reduction , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
6.
Chemotherapy ; 59(3): 192-9, 2013.
Article in English | MEDLINE | ID: mdl-24335094

ABSTRACT

BACKGROUND/AIM: Enforced expression of the connexin (Cx) 32 gene, a member of the gap junction gene family and a tumor suppressor gene in human renal cell carcinoma (RCC), enhanced vinblastine (VBL)-induced cytotoxicity in RCC cells due to suppression of multidrug resistance 1 (MDR1) expression. Furthermore, in RCC the Cx32 gene is silenced by hypermethylation of CpG islands in a promoter region of the Cx gene. In this study, we investigated if the green tea polyphenol epigallocatechin-3-gallate (EGCG) could enhance susceptibility of RCC cells (Caki-1, a human metastatic RCC cell) to VBL. METHODS: The effects of EGCG on Caki-1 cells were estimated by WST-1 (cell viability), real-time RT-PCR (mRNA level) and immunoblotting (protein level). We estimated the methylation status in the promoter region of the Cx32 gene in RCC cells by methylation-specific PCR. Each protein function was inhibited by small interfering RNA (siRNA) and specific inhibitors. RESULTS: The EGCG treatment elicited significant upregulation of Cx32 in Caki-1 cells, and the induction of the Cx led to the suppression of MDR1 mRNA expression through inactivation of Src and subsequent activation of c-Jun NH2-terminal kinase (JNK). Chemical sensitivity to VBL in Caki-1 cells was increased by EGCG pretreatment, and this effect was abrogated by siRNA-mediated knockdown of Cx32. CONCLUSION: This study suggests that the restoration of Cx32 by EGCG pretreatment improves chemical tolerance on VBL in Caki-1 cells via the inactivation of Src and the activation of JNK.


Subject(s)
Catechin/analogs & derivatives , Cell Survival/drug effects , Connexins/genetics , Vinblastine/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Catechin/pharmacology , Cell Line, Tumor , Connexins/antagonists & inhibitors , Connexins/metabolism , DNA Methylation , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Up-Regulation/drug effects , Gap Junction beta-1 Protein
7.
J Appl Toxicol ; 33(9): 924-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22729658

ABSTRACT

Coleus forskohlii root extract (CFE) represented by its bioactive constituent 'forskolin' is popularly used as a natural weight-lowering product, but the association of its use with liver-related risks is very limited. In the present study, the effect of standardized CFE with 10% forskolin on liver function of mice was examined. Mice were given 0-5% CFE in an AIN93G-based diet for 3-5 weeks. Food intake, body weights, relative organ weights and liver marker enzymes [aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP)] combined with histophatological analysis were assessed. CFE (0-0.5%) only had minimal effects on food intake and body weight whereas a significant difference was observed in mice receiving the highest dose (5% CFE). The extract 0.05-5% dose-dependently decreased visceral fat weight by between 16% and 63%, and a dose-dependent several folds increase was observed in liver weights and plasma AST, ALT and ALP activities with quick onset apparent after only 1 week of 0.5% CFE intake. The hepatic effect persisted throughout the 3-weeks course but was restored towards normalization within 1 week after withdrawal of treatment. Liver histology of mice fed 0.5% CFE for 3 weeks showed hepatocyte hypertrophy and fat deposition. In contrast, none of the hepatic responses measured were altered when mice were given a diet containing pure forskolin alone at the dose corresponding to its content in 0.5% CFE. The present study clearly indicated that forskolin was not involved in the CFE-induced hepatotoxicity and was caused by other unidentified constituents in CFE which warrants further studies.


Subject(s)
Coleus/chemistry , Liver/drug effects , Liver/pathology , Plant Extracts/adverse effects , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Body Weight , Colforsin/adverse effects , Colforsin/toxicity , Diet , Dose-Response Relationship, Drug , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Mice , Mice, Inbred ICR , Organ Size/drug effects , Plant Extracts/administration & dosage , Plant Roots/chemistry
8.
J Vet Med Sci ; 85(7): 721-726, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37225449

ABSTRACT

This report described the differentiation induction of canine oral mucosal melanoma (OMM) cells by resveratrol. Exposure of canine OMM cells to resveratrol (maximum dose: 50 µM and treatment period: 72 hr) induced differentiating features like melanocytes, and enhanced chemosensitivity against cisplatin, but alone had no influence on cell viability. Additionally, resveratrol significantly enhanced mRNA expression of key melanoma differentiation markers such as microphthalmia-associated transcription factor (MITF). Of several inhibitors against mitogen-activated protein kinase subtypes, only the c-Jun N-terminal kinase (JNK) inhibitor, SP600125, induced melanocyte-like morphological change and enhanced MITF mRNA expression. Furthermore, resveratrol also suppressed JNK activation in OMM cells by approximately 33%. Overall, these findings suggest that resveratrol induces differentiation in canine OMM cells, due to the inhibition of JNK signaling.


Subject(s)
Dog Diseases , Melanoma , Animals , Dogs , Resveratrol/pharmacology , Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/veterinary , RNA, Messenger/metabolism , Dog Diseases/drug therapy
9.
Phytother Res ; 24(9): 1317-21, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20127663

ABSTRACT

Resistance to chemotherapy (chemoresistance) is a serious problem in malignant mesothelioma, a highly aggressive neoplasm. Gamma-tocotrienol (gamma-T3) can sensitize various cancerous cells to chemotherapeutic agents by inhibiting pathways that lead to treatment resistance. In this study, we investigated the modulating effect of tocotrienol-rich fraction (TRF) from rice bran, which is abundant in gamma-T3, on chemoresistance in human MM H28 cells. TRF treatment caused a marked reduction in the viability of H28 cells in a dose-dependent manner, while cisplatin treatment had no effect on the cells, indicating that H28 cells are resistant to cisplatin. A significant increase in cytotoxicity was observed in H28 cells treated with TRF, and this effect was enhanced by the combination treatment with cisplatin. The cytotoxic effect was closely related to the inhibition of phosphatidylinositol 3-kinase (PI3K)-AKT signaling. Inactivation of Akt signaling by TRF or the combination with cisplatin mitigated cisplatin-induced activation of Akt, resulting in reducing the chemoresistance H28 cells to cisplatin. Reduced cell viability and attenuated chemoresistance of the H28 cells against cisplatin were also observed following the use of a PI3K inhibitor, LY294002. These results suggest that the combination therapy of cisplatin with TRF is a plausible strategy for achieving tolerance for the chemotherapeutic agent in MM therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Chromans/pharmacology , Cisplatin/therapeutic use , Mesothelioma/drug therapy , Oryza/chemistry , Plant Extracts/pharmacology , Vitamin E/analogs & derivatives , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Chromans/therapeutic use , Chromones/pharmacology , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Drug Therapy, Combination , Humans , Mesothelioma/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Seeds , Signal Transduction/drug effects , Vitamin E/pharmacology , Vitamin E/therapeutic use
10.
Biochem Biophys Res Commun ; 365(4): 875-81, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18042466

ABSTRACT

We have previously reported that a redox-silent analogue of alpha-tocotrienol (T3), 6-O-carboxypropyl-alpha-tocotrienol (T3E) shows more potential anti-carcinogenic property than T3 in a lung cancer cell (A549 cell). However, the mechanisms by which T3E exerts its potential anti-carcinogenic effect is still unclear. As tumor malignancy is associated with hypoxia adaptation, in this study, we examined whether T3E could suppress survival and invasion in A549 cells under hypoxia. Hypoxia treatment drastically-induced activation of the protein tyrosine kinase, Src, and its regulated signaling required for hypoxia adaptation of A549 tumor cells. The survival and invasion capacity of the tumor cells under hypoxia was suppressed by T3E via the inactivation of Src. More specifically, T3E-dependent inhibition of Src-induced Akt activation contributed to suppression of cell survival under hypoxia, and the reduction of fibrinolytic factors such as plasminogen activator-1(PAI-1) via the decrease of hypoxia-inducible factor-2alpha by T3E led to inhibition of hypoxic invasion. Overall these results suggest that T3E suppresses hypoxia adaptation of A549 cells by the inhibition in hypoxia-induced activation of Src signaling.


Subject(s)
Cell Survival/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oxygen/metabolism , Tocotrienols/administration & dosage , Adaptation, Physiological/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Humans , Oxidation-Reduction/drug effects
11.
Anticancer Res ; 38(3): 1391-1399, 2018 03.
Article in English | MEDLINE | ID: mdl-29491063

ABSTRACT

BACKGROUND/AIM: A hallmark of the progression of prostate cancer to advanced disease is the acquisition of androgen-independent growth. This malignant phenotype is characterized by resistance to conventional treatments and predisposes to formation of hypoxic regions containing stem-like cancer cells. Unfortunately, an effective therapy to target prostate cancer stem cells under hypoxia has not yet been established. In this report, we studied whether δ-tocotrienol (T3), a vitamin E family member that has exhibited the most potent anti-cancer activity, could suppress the survival of prostate cancer stem-like cells. MATERIALS AND METHODS: PC3 stem-like cells were isolated from PC3 parental cells using a three-dimensional culture system. The stemness of the isolated PC3 stem-like cells was confirmed by evaluation of resistance to an anticancer agent (docetaxel) and tumor formation capacity in a xenograft model. The effects of δ-T3 on PC3 stem-like cells under a hypoxia condition were examined by WST-8 (cell viability), real-time reverse transcription-polymerase chain reaction (PCR) and western blotting. RESULTS: δ-T3 demonstrated a cytotoxic effect on prostate cancer stem-like cells in a dose dependent manner and a reduction in the protein levels of hypoxia-inducible factor (HIF)-1α and HIF-2α. Additionally, a specific inhibitor toward HIF-1α induced cytotoxicity on PC3 cells, but selective inhibition of HIF-2α had no effect. CONCLUSION: Overall, these results suggest that δ-T3 could inhibit the survival of prostate cancer stem-like cells under hypoxia, primarily through the inactivation of HIF-1α signaling.


Subject(s)
Adaptation, Physiological/drug effects , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , Vitamin E/analogs & derivatives , Adaptation, Physiological/genetics , Animals , Cell Hypoxia , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia , Male , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tumor Cells, Cultured , Vitamin E/pharmacology , Vitamins/pharmacology , Xenograft Model Antitumor Assays
12.
Cancer Lett ; 253(2): 249-57, 2007 Aug 18.
Article in English | MEDLINE | ID: mdl-17343982

ABSTRACT

Bowman-Birk protease inhibitor (BBI) from soybean acts as a potential chemopreventive agent in several types of tumors. However, the mechanism is still unclear. The present study was undertaken to estimate a mechanism of BBI-dependent negative growth control of human osteosarcoma cell (U2OS cell). BBI had negative growth control of the cells via induction of connexin (Cx) 43, a tumor suppressor gene in U2OS cells. This negative growth control by BBI was abrogated under down-regulation of Cx43 induced by a Cx43 antisense nucleotide treatment. It was also found that the BBI-dependent induction of Cx43 was due to elevation of Cx43 mRNA and stabilization of Cx43 protein. Especially, BBI-dependent inhibition of chymotrypsin-like activity in proteasome contributed to stabilization of Cx43 protein. These results suggest that a major negative growth effect of BBI is based on the restoration of Cx43 expression in U2OS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Connexin 43/metabolism , Osteosarcoma/metabolism , Protease Inhibitors/pharmacology , Trypsin Inhibitor, Bowman-Birk Soybean/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Connexin 43/genetics , Humans , Up-Regulation
13.
J Nutr Sci Vitaminol (Tokyo) ; 63(5): 349-354, 2017.
Article in English | MEDLINE | ID: mdl-29225320

ABSTRACT

Tocotrienols (T3s) and tocopherols (Tocs) are both members of the vitamin E family. It is known that δ-tocotrienol (δ-T3) has displayed the most potent anti-cancer activity amongst the tocotrienols. On the other hand, γ-tocopherol (γ-Toc) is reported to have a protective effect against prostate cancer. Therefore, we investigated whether the combination of γ-Toc and δ-T3 could strengthen the inhibitory effect of δ-T3 on prostate cancer cell growth. In this study the effect of combined δ-T3 (annatto T3 oil) and γ-Toc (Tmix, γ-Toc-rich oil) therapy was assessed against human androgen-dependent prostate cancer cells (LNCaP). We found that combined treatment of δ-T3 (10 µM) and γ-Toc (5 µM) resulted in reinforced anti-prostate cancer activity. Specifically, cell cycle phase distribution analysis revealed that in addition to G1 arrest caused by the treatment with δ-T3, the combination of δ-T3 with γ-Toc induced G2/M arrest. Enhanced induction of apoptosis by the combined treatment was also observed. These findings indicate that combination of δ-T3 and γ-Toc significantly inhibits prostate cancer cell growth due to the simultaneous cell cycle arrest in the G1 phase and G2/M phase.


Subject(s)
Anticarcinogenic Agents/metabolism , Antineoplastic Agents, Phytogenic/agonists , Apoptosis , Chromans/agonists , Prostatic Neoplasms/metabolism , Vitamin E/analogs & derivatives , Anticarcinogenic Agents/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Bixaceae/metabolism , Carotenoids/agonists , Carotenoids/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromans/metabolism , G1 Phase , G2 Phase , Humans , Male , Osmolar Concentration , Plant Extracts/agonists , Plant Extracts/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Vitamin E/agonists , Vitamin E/metabolism
14.
Oncol Rep ; 16(5): 1149-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016607

ABSTRACT

Recent evidence suggests that a member of the gap junction protein family, connexin (Cx) 32, acts as a tumor suppressor gene against lung adenocarcinoma. However, the precise mechanism remains unclear. In this study, we tried to explore the mechanism for the Cx32-dependent tumor-suppressive effect in lung adenocarcinoma. To perform this study, we established a stable clone of the human lung adenocarcinoma cell line, A549 in which the Cx32 gene was expressed. Cx32 expression in A549 cells reduced anchorage-independent growth and development of tumors in a xenograft model. Additionally, Cx32 induced contact inhibition of growth and reduced invasive activity in A549 cells. The tumor-suppressive effects of Cx32 depended on the inhibition of Src activity. These events were confirmed by an Src inhibitor (PP1) and siRNA for Cx32. These results suggest that the Cx32-dependent tumor-suppressive effect in A549 cells is explained by the inhibition of Src activity.


Subject(s)
Adenocarcinoma/metabolism , Connexins/biosynthesis , Lung Neoplasms/metabolism , src-Family Kinases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Connexins/genetics , Genes, Tumor Suppressor , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Transfection , Transplantation, Heterologous , src-Family Kinases/antagonists & inhibitors , Gap Junction beta-1 Protein
15.
Oncol Rep ; 15(5): 1359-65, 2006 May.
Article in English | MEDLINE | ID: mdl-16596211

ABSTRACT

Connexin (Cx) genes exert negative growth effects on tumor cells with certain cell specificity. We have recently reported that Cx32 acts as a tumor suppressor gene in a metastatic renal cancer cell line (Caki-1) due to the inactivation of Src. In line with the previous study, here we investigated if an Src family inhibitor (PP1) could enhance the tumor-suppressive effect of Cx32 in Caki-1 cells from human metastatic renal cell carcinoma. We examined the difference in the cytotoxic effect of PP1 on two cell clones, Cx32-transfected Caki-1 cells (Caki-1T) and mock-transfected Caki-1 cells (Caki-1W), in vitro as well as in vivo. PP1 showed more cytotoxic effect on Caki-1T than on Caki-1W at lower doses. This reinforcement was also observed in a xenograft model of nude mice. The in vitro reinforcement of the cytotoxic effect depended not only on control of cell-cycle transition but also on the induction of apoptosis, and the occurrence of the event was mostly caused by potential inhibition of Src activity in Caki-1T. Also, under a hypoxic condition, which is a typical environment of tumor tissue, Cx32 suppressed hypoxia-induced Src activation, and PP1 enhanced cytotoxicity in Caki-1T. These results suggest that, in addition to the Cx32-dependent tumor-suppressive effect, the concomitant inhibition of Src by PP1 is an effective procedure to induce a cytotoxic effect in Caki-1 cells.


Subject(s)
Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Connexins/metabolism , Enzyme Inhibitors/therapeutic use , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Cycle/drug effects , Cell Hypoxia , Connexins/genetics , Enzyme Activation/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins pp60(c-src)/metabolism , Transfection , Tumor Cells, Cultured , Gap Junction beta-1 Protein
16.
FEBS Lett ; 579(17): 3829-36, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15978581

ABSTRACT

It has been assumed that prostaglandin (PG)I2 signaling contributes to the negative growth control of lung cancer cells; however, the mechanism remains unresolved. PGI2 functions through a cell surface G protein-coupled receptor (prostaglandin I2-binding receptor, IP) and also exerts an effect by interacting with a nuclear hormone receptor, peroxisome proliferator-activated receptor delta (PPARdelta). We found that PPARdelta was a key molecule of PGI2 signaling to give negative growth control of lung cancer cells (A549), using carbarprostacyclin, a PGI2 agonist for IP and PPARdelta, and L-165041, a PPARdelta agonist. Furthermore, PPARdelta-induced cell growth control was reinforced by the inhibition of cyclooxygenase. These results suggest that PPARdelta activation under the suppression of PG synthesis is important to regulate lung cancer cell growth.


Subject(s)
Epoprostenol/metabolism , Lung Neoplasms/metabolism , PPAR delta/metabolism , Receptors, Epoprostenol/metabolism , Apoptosis , Down-Regulation , Epoprostenol/agonists , Epoprostenol/pharmacology , Humans , Lung Neoplasms/drug therapy , PPAR delta/agonists , PPAR delta/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Epoprostenol/genetics , Signal Transduction , Tumor Cells, Cultured , Up-Regulation
17.
J Nutr Sci Vitaminol (Tokyo) ; 61(6): 497-501, 2015.
Article in English | MEDLINE | ID: mdl-26875492

ABSTRACT

Prostate cancer is one of the most frequently occurring cancers and often acquires the potential of androgen-independent growth as a malignant phenotype. Androgen-independent prostate cancer has severe chemoresistance towards conventional chemotherapeutic agents, so a new treatment approach is required for curing such prostate cancer. In this context, the present study was undertaken to check if annatto tocotrienol (main component δ-tocotrienol) could suppress cell growth in human prostate cancer (PC3, androgen-independent type) cells via the inhibition of Src and Stat3. The tocotrienol showed cytotoxic effects on PC3 cells in a dose-dependent manner, and the effect depended on G1 arrest in the cell cycle and subsequent induction of apoptosis. In a cytotoxic dose, the tocotrienol suppressed cellular growth via the simultaneous inhibition of Src and Stat3. Similarly, the treatment combination of both Src and Stat3 inhibitors induced cytotoxic effects in PC3 cells in an additive manner compared to each by itself. With respect to cell cycle regulation and the induction of apoptosis, the combination treatment showed a similar effect to that of the tocotrienol treatment. These results suggest that annatto tocotrienol effectively induces cytotoxicity in androgen-independent prostate cancer cells via the suppression of Src and Stat3.


Subject(s)
Antineoplastic Agents/therapeutic use , Bixaceae/chemistry , Carotenoids/chemistry , Plant Extracts/chemistry , Prostatic Neoplasms/drug therapy , Tocotrienols/therapeutic use , Vitamins/therapeutic use , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Genes, src , Humans , Male , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Tocotrienols/pharmacology , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Vitamin E/therapeutic use , Vitamins/pharmacology
18.
Anticancer Agents Med Chem ; 13(3): 496-501, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22721395

ABSTRACT

Vitamins are prominent among natural or endogenous compounds that are considered to be beneficial for both prevention and therapy of various human ailments. The vitamin E group of compounds composed of tocopherol and tocotrienol isoforms, has been subsequently proven to have health benefits including antioxidant and related protective properties. However, individual isoforms exhibit a wide-range of antioxidant potencies. Tocotrienol (T3) displays powerful anticancer activity that is often not exhibited by tocopherols, by modulating multiple intracellular signaling pathways associated with tumor cell proliferation and survival. The anticancer effect of T3 remains not fully understood but generally is mediated independently of its antioxidant activity. Further we have synthesized a new redox-inactive analogue of T3, 6-O-carboxypropyl-α-tocotrienol (T3E) showing considerable promise for stronger anticancer potency than its mother compound. In this mini-review, we particularly focus upon the anticancer action of the above active components of vitamin E and describe current research on the anticancer effects of T3 irrespective of antioxidant activity.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Tocotrienols/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Half-Life , Humans , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Oxidation-Reduction , Signal Transduction/drug effects , Tocotrienols/chemical synthesis , Tocotrienols/pharmacokinetics
19.
Oncol Rep ; 28(5): 1889-93, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22948717

ABSTRACT

The constitutive activation of the Src family kinases (SFKs) has been established as a poor prognostic factor in malignant mesothelioma (MM), however, the family member(s) which contribute to the malignancy have not been defined. This study aimed to identify the SFK member(s) contributing to cell growth using RNA interference in various MM cell lines. Silencing of Yes but not of c-Src or Fyn in MM cells leads to cell growth suppression. This suppressive effect caused by Yes silencing mainly depends on G1 cell cycle arrest and partly the induction of apoptosis. Also, the knockout of Yes induces the inactivation of ß-catenin signaling and subsequently decreases the levels of cyclin D necessary for G1-S transition in the cell cycle. In addition, Yes knockout has less effect on cell growth suppression in ß-catenin-deficient H28 MM cells compared to other MM cells which express the catenin. Overall, we conclude that Yes is a central mediator for MM cell growth that is not shared with other SFKs such as c-Src.


Subject(s)
Mesothelioma/metabolism , Mesothelioma/pathology , Proto-Oncogene Proteins c-yes/metabolism , Apoptosis/genetics , Cell Division , Cell Line, Tumor , Cell Proliferation , Cyclin D/biosynthesis , G1 Phase Cell Cycle Checkpoints , Humans , Mesothelioma/genetics , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-yes/genetics , RNA Interference , RNA, Small Interfering , beta Catenin/metabolism , src-Family Kinases/genetics
20.
Food Chem Toxicol ; 50(3-4): 750-5, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22178802

ABSTRACT

Coleus forskohlii root extract (CFE) is popular for use as a weight loss dietary supplement. In this study, the influence of standardized CFE containing 10% active component forskolin on the hepatic drug metabolizing system was investigated to evaluate the safety through its drug interaction potential. Male ICR mice were fed AIN93G-based diets containing 0-5% CFE or 0.05% pure forskolin for 2-3 weeks. Intake of two different sources of 0.5% CFE significantly increased the relative liver weight, total content of hepatic cytochrome P450 (CYP) and induced CYPs (especially 2B, 2C, 3A types) and glutathione S-transferase (GST) activities. CFE significantly increased mRNA expression of CYPs and GST with dose related responses. However, unlike the CFE, intake of 0.05% pure forskolin was found to be associated with only weak induction in CYP3A and GST activities with no significant increases in relative liver weight, total hepatic content or other CYPs activities. The inductions of CYPs and GST by CFE were observed at 1 week of feeding and rapidly recovered by discontinuation of CFE. These results indicated the induction potential of CFE on CYPs, and that this effect was predominantly due to other, as yet unidentified constituents, and not forskolin contained in CFE.


Subject(s)
Coleus/chemistry , Cytochrome P-450 Enzyme System/biosynthesis , Microsomes, Liver/enzymology , Plant Extracts/pharmacology , Animals , Base Sequence , DNA Primers , Enzyme Induction , Glutathione Transferase/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred ICR , Organ Size/drug effects , Plant Roots/chemistry , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL