Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Biomolecules ; 10(11)2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33172112

RESUMEN

Lung cancer is the major cause of cancer-associated death worldwide, and development of new therapeutic drugs is needed to improve treatment outcomes. Three-dimensional (3D) tumorspheroids offer many advantages over conventional two-dimensional cell cultures due to the similarities to in vivo tumors. We found that isoharringtonine, a natural product purified from Cephalotaxus koreana Nakai, significantly inhibited the growth of tumorspheroids with NCI-H460 cells in a dose-dependent manner and induced apoptotic cell death in our 3D cell culture system. On the other hand, A549 tumorspheroids displayed low sensitivity to isoharringtonine-induced apoptosis. Nuclear receptor subfamily 4 group A member 1 (NR4A1) is an orphan nuclear receptor known to regulate proliferation and apoptosis of cancer cells. We observed that knockdown of NR4A1 dramatically increased isoharringtonine-induced cancer cell death in A549 tumorspheroids by activating the intrinsic apoptosis pathway. Furthermore, treatment with combined isoharringtonine and iNR4A1 significantly inhibited multivulva formation in a Caenorhabditis elegans model and tumor development in a xenograft mouse model. Taken together, our data suggest that isoharringtonine is a potential natural product for treatment of non-small cell lung cancers, and inhibition of NR4A1 sensitizes cancer cells to anti-cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Harringtoninas/farmacología , Neoplasias Pulmonares/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Mol Med (Berl) ; 98(11): 1591-1602, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32901343

RESUMEN

Nitidine chloride (NC) was recently reported to exhibit a wide range of pharmacological properties for several diseases, including cancer. Here we report for the first time that NC is a potential therapeutic agent for mucoepidermoid carcinoma (MEC) occurring in the head and neck because it suppresses X chromosome-linked inhibitor of apoptosis protein (XIAP) in human MEC in vitro and in vivo. The antitumor effects of NC were evaluated by trypan blue exclusion assay, western blotting, live/dead assay, 4',6-diamidino-2-phenylindole (DAPI) staining, human apoptosis antibody array, immunofluorescence staining, immunohistochemistry, small interfering RNA assay, transient transfection of XIAP overexpression vector, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and histopathological examination of organs. NC inhibited cell viability and induced caspase-dependent apoptosis in vitro. A human apoptosis antibody array assay showed that XIAP is suppressed by NC treatment. XIAP was overexpressed in oral squamous cell carcinoma (OSCC) tissues that arose from the head and neck, and high XIAP expression was correlated with poor prognosis in OSCC patients. XIAP depletion significantly increased apoptosis, and ectopic XIAP overexpression attenuated the apoptosis induced by NC treatment. NC suppressed tumor growth in vivo at a dosage of 5 mg/kg/day. The number of TUNEL-positive cells increased and the protein expression of XIAP was consistently downregulated in NC-treated tumor tissues. In addition, NC caused no histopathological changes in the liver or kidney. These findings provide new insights into the mechanism of action underlying the anticancer effects of NC and demonstrate that NC is a promising therapeutic agent for the treatment of human MEC of the head and neck. KEY MESSAGES: • Nitidine chloride induces caspase-dependent apoptosis in MEC of the head and neck. • High XIAP expression correlates with poor prognosis of OSCC patients. • Nitidine chloride suppresses tumor growth in vivo without any systemic toxicities. • Targeting XIAP is a novel chemotherapeutic strategy for MEC of the head and neck.


Asunto(s)
Antineoplásicos/farmacología , Benzofenantridinas/farmacología , Biomarcadores de Tumor , Carcinoma Mucoepidermoide/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Terapia Molecular Dirigida , Proteína Inhibidora de la Apoptosis Ligada a X/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Carcinoma Mucoepidermoide/tratamiento farmacológico , Carcinoma Mucoepidermoide/etiología , Carcinoma Mucoepidermoide/patología , Línea Celular Tumoral , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
3.
Int J Biol Sci ; 16(11): 1901-1916, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32398958

RESUMEN

Ovarian cancer is a common gynecological cancer that is found worldwide. Class III histone deacetylase (HDAC) inhibitors, a new class of anticancer agents, induce autophagy in various human cancer cells. The aim of the present study was to investigate the antitumor activity of MHY2245, a new synthetic SIRT inhibitor, on human ovarian cancer cells. We found that MHY2245 exhibited potent cytotoxicity to SKOV3 cells in a time- and concentration-dependent manner. The cytotoxicity of MHY2245 (IC50=0.32 µM) was higher than that of doxorubicin (DOX, IC50=1.38µM) against SKOV3 cells. MHY2245 significantly inhibited SIRT1 enzyme activity, reduced the expression of SIRT1, increased cell cycle arrest at G2/M phase, and induced apoptotic cell death in SKOV3 cells via expression of cytochrome c, cleaved-PARP, cleaved caspase-3, and Bax. This might be associated with blocking of the pyruvate kinase M2 (PKM2)/mTOR pathway. MHY2245 also inhibited tumor growth and reduced tumor size when SKOV3 cells were transplanted into nude mice. Our results indicate that MHY2245 exerts antitumor activity against ovarian cancer cells by blocking the PKM2/mTOR pathway. We suggest that MHY2245 is a promising anticancer agent that disrupts ovarian cancer cell metabolism.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Proteínas Portadoras/metabolismo , Metabolismo Energético/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Sirtuina 1/antagonistas & inhibidores , Hormonas Tiroideas/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Metabolismo Energético/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Neoplasias Experimentales , Neoplasias Ováricas , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Hormonas Tiroideas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión a Hormona Tiroide
4.
Int J Mol Sci ; 20(22)2019 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-31717694

RESUMEN

Emerging evidence indicates that the activity of pyruvate kinase M2 (PKM2) isoform is crucial for the survival of tumor cells. However, the molecular mechanism underlying the function of PKM2 in renal cancer is undetermined. Here, we reveal the overexpression of PKM2 in the proximal tubule of renal tumor tissues from 70 cases of patients with renal carcinoma. The functional role of PKM2 in human renal cancer cells following small-interfering RNA-mediated PKM2 knockdown, which retarded 786-O cell growth was examined. Targeting PKM2 affected the protein kinase B (AKT)/mechanistic target of the rapamycin 1 (mTOR) pathway, and downregulated the expression of glycolytic enzymes, including lactate dehydrogenase A and glucose transporter-1, and other downstream signaling key proteins. PKM2 knockdown changed glycolytic metabolism, mitochondrial function, adenosine triphosphate (ATP) level, and intracellular metabolite formation and significantly reduced 786-O cell migration and invasion. Acridine orange and monodansylcadaverine staining, immunocytochemistry, and immunoblotting analyses revealed the induction of autophagy in renal cancer cells following PKM2 knockdown. This is the first study to indicate PKM2/AKT/mTOR as an important regulatory axis mediating the changes in the metabolism of renal cancer cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/patología , Proteínas Portadoras/metabolismo , Movimiento Celular , Proliferación Celular , Neoplasias Renales/patología , Proteínas de la Membrana/metabolismo , Hormonas Tiroideas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Proteínas Portadoras/genética , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Pronóstico , Hormonas Tiroideas/genética , Células Tumorales Cultivadas , Proteínas de Unión a Hormona Tiroide
5.
Cell Physiol Biochem ; 52(6): 1535-1552, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31135122

RESUMEN

BACKGROUND/AIMS: Pyruvate kinase M2 (PKM2) is essential for aerobic glycolysis. Although high PKM2 expression is observed in various cancer tissues, its functional role in cancer metabolism is unclear. Here, we investigated the role of PKM2 in regulating autophagy and its associated pathways in prostate cancer cells. METHODS: Immunohistochemistry was performed to compare the expression level of PKM2 in prostate cancer patients and normal human, whereas expression of PKM2 in several cell lines was also examined by using western blot. PKM2 expression was silenced using various small interfering RNAs (siRNAs). Cell viability was examined using IncuCyte ZOOM™ live cell imaging system. Western blotting and immunofluorescence were performed to investigate the PKM2 knockdown on other cellular signaling molecules. Acridine orange and Monodansylcadaverine staining was performed to check effect of PKM2 knockdown on autophagy induction. High performance thin layer chromatography was carried out to quantify the level of different cellular metabolites (pyruvate and lactate). Colony formation assay was performed to determine the ability of a cells to form large colonies. RESULTS: PKM2 was highly expressed in prostate cancer patients as compared to normal human. PKM2 siRNA-transfected prostate cancer cells showed significantly reduced viability. Acridine orange, Monodansylcadaverine staining and western blotting analysis showed that PKM2 downregulation markedly increased autophagic cell death. Results of western blotting analysis showed that PKM2 knockdown affected protein kinase B/mechanistic target of rapamycin 1 pathway, which consequently downregulated the expression of glycolytic enzymes lactate dehydrogenase A and glucose transporter 1. Knockdown of PKM2 also reduced the colony formation ability of human prostate cancer cell DU145. CONCLUSION: To the best of our knowledge, this is the first study to show that PKM2 inhibition alters prostate cancer cell metabolism and induces autophagy, thus providing new perspectives for developing PKM2-targeting anticancer therapies for treating prostate cancer.


Asunto(s)
Autofagia , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Quinasa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Masculino , Neoplasias de la Próstata/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal
6.
Cells ; 8(3)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30866413

RESUMEN

Chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI) is an orphan receptor and member of the nuclear receptor superfamily. Among a series of methylene substituted diindolylmethanes (C-DIMs) containing substituted phenyl and heteroaromatic groups, we identified 1,1-bis(3'-indolyl)-1-(4-pyridyl)-methane (DIM-C-Pyr-4) as an activator of COUP-TFI. Structure activity studies with structurally diverse heteroaromatic C-DIMs showed that the pyridyl substituted compound was active and the 4-pyridyl substituent was more potent than the 2- or 3-pyridyl analogs in transactivation assays in breast cancer cells. The DIM-C-Pyr-4 activated chimeric GAL4-COUP-TFI constructs containing full length, C- or N-terminal deletions, and transactivation was inhibited by phosphatidylinositol-3-kinase and protein kinase A inhibitors. However, DIM-C-Pyr-4 also induced transactivation and interactions of COUP-TFI and steroid receptor coactivators-1 and -2 in mammalian two-hybrid assays, and ligand-induced interactions of the C-terminal region of COUP-TFI were not affected by kinase inhibitors. We also showed that DIM-C-Pyr-4 activated COUP-TFI-dependent early growth response 1 (Egr-1) expression and this response primarily involved COUP-TFI interactions with Sp3 and to a lesser extent Sp1 bound to the proximal region of the Egr-1 promoter. Modeling studies showed interactions of DIM-C-Pyr-4 within the ligand binding domain of COUP-TFI. This report is the first to identify a COUP-TFI agonist and demonstrate activation of COUP-TFI-dependent Egr-1 expression.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Indoles/farmacología , Animales , Factor de Transcripción COUP I/química , Línea Celular Tumoral , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Ligandos , Ratones , Modelos Moleculares , Co-Represor 2 de Receptor Nuclear/metabolismo , Factores de Transcripción Sp/metabolismo
7.
Int J Biol Sci ; 15(4): 800-811, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906211

RESUMEN

Hepatic fibrosis, characterized by persistent deposition of extracellular matrix (ECM) proteins, occurs in most types of chronic liver disease. The prevention of liver damage using extract of Dendropanax morbifera has been widely studied, but its molecular mechanism on the therapeutic efficacy of hepatic fibrosis is unclear. The aim of this study was to assess whether aquatic extract (DM) of D. morbifera ameliorates thioacetamide (TAA)-induced hepatic fibrosis. Hepatic fibrosis was induced by an intraperitoneal (i.p.) injection (150 mg/kg, twice per week) of TAA for 6 weeks. DM (50 mg/kg/day) or silymarin (50 mg/kg/day) was administered daily for 6 weeks. DM markedly reduced serum AST, ALT, ALP, and r-GTP in TAA-treated rats. DM significantly ameliorated the total glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT) activity in TAA-treated rats. In particular, DM significantly reduced expression of α-SMA, type I collagen, vimentin, TGF-ß1 and p-Smad2/3 in hepatic fibrosis rats. The protective effects of DM on progression of hepatic fibrosis were clearly shown by detecting 4-hydroxyproline concentration and histopathological examination in the liver. Therefore, our data suggest that DM dramatically prevented hepatic fibrosis by inhibiting oxidative stress and the TGF-ß1/Smads signaling pathways.


Asunto(s)
Araliaceae/química , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Proteína Smad2/metabolismo , Tioacetamida/toxicidad , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Western Blotting , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo
8.
Cells ; 8(2)2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30754676

RESUMEN

The CCAAT/enhancer-binding protein ß (C/EBPß) is a transcription factor that regulates cellular proliferation, differentiation, apoptosis and tumorigenesis. Although the pro-oncogenic roles of C/EBPß have been implicated in various human cancers, how it contributes to tumorigenesis or tumor progression has not been determined. Immunohistochemistry with human non-small cell lung cancer (NSCLC) tissues revealed that higher levels of C/EBPß protein were expressed compared to normal lung tissues. Knockdown of C/EBPß by siRNA reduced the proliferative capacity of NSCLC cells by delaying the G2/M transition in the cell cycle. In C/EBPß-knockdown cells, a prolonged increase in phosphorylation of cyclin dependent kinase 1 at tyrosine 15 (Y15-pCDK1) was displayed with simultaneously increased Wee1 and decreased Cdc25B expression. Chromatin immunoprecipitation (ChIP) analysis showed that C/EBPß bound to distal promoter regions of WEE1 and repressed WEE1 transcription through its interaction with histone deacetylase 2. Treatment of C/EBPß-knockdown cells with a Wee1 inhibitor induced a decrease in Y15-pCDK1 and recovered cells from G2/M arrest. In the xenograft tumors, the depletion of C/EBPß significantly reduced tumor growth. Taken together, these results indicate that Wee1 is a novel transcription target of C/EBPß that is required for the G2/M phase of cell cycle progression, ultimately regulating proliferation of NSCLC cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular , Fase G2 , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , División Celular/efectos de los fármacos , División Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Fase G2/efectos de los fármacos , Fase G2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones Desnudos , Persona de Mediana Edad , Modelos Biológicos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Pirazoles/farmacología , Pirimidinonas/farmacología , Transcripción Genética/efectos de los fármacos
9.
Lab Invest ; 99(8): 1157-1172, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30700846

RESUMEN

Doxorubicin is a widely used DNA damage-inducing anti-cancer drug. However, its use is limited by its dose-dependent side effects, such as cardiac toxicity. Cholesterol-lowering statin drugs increase the efficacy of some anti-cancer drugs. Cholesterol is important for cell growth and a critical component of lipid rafts, which are plasma membrane microdomains important for cell signaling. 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMG-CR) is a critical enzyme in cholesterol synthesis. Here, we show that doxorubicin downregulated HMG-CR protein levels and thus reduced levels of cholesterol and lipid rafts. Cholesterol addition attenuated doxorubicin-induced cell death, and cholesterol depletion enhanced it. Reduction of HMG-CR activity by simvastatin, a statin that acts as an HMG-CR inhibitor, or by siRNA-mediated HMG-CR knockdown enhanced doxorubicin cytotoxicity. Doxorubicin-induced HMG-CR downregulation was associated with inactivation of the EGFR-Src pathway. Furthermore, a high-cholesterol-diet attenuated the anti-cancer activity of doxorubicin in a tumor xenograft mouse model. In a multivulva model of Caenorhabditis elegans expressing an active-EGFR mutant, doxorubicin decreased hyperplasia more efficiently in the absence than in the presence of cholesterol. These data indicate that EGFR/Src/HMG-CR is a new pathway mediating doxorubicin-induced cell death and that cholesterol control could be combined with doxorubicin treatment to enhance efficacy and thus reduce side effects.


Asunto(s)
Antineoplásicos/farmacología , Doxorrubicina/farmacología , Receptores ErbB/metabolismo , Hidroximetilglutaril-CoA Reductasas/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo , Animales , Caenorhabditis elegans , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biochim Biophys Acta Mol Cell Res ; 1866(5): 761-772, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30710564

RESUMEN

Transforming growth factor-beta (TGF-ß) is a known tumor suppressor, which also exerts a tumor promoting activity at an advanced stage of cancer. Previously, we reported that expression of interferon consensus sequence-binding protein (ICSBP), also known as interferon regulatory factor-8, is positively correlated with TGF-ß type I receptor (TGF-ß RI) expression in osteosarcoma patient tissues. In this study, we demonstrated that ICSBP upregulated TGF-ß RI and induced epithelial-to-mesenchymal transition-like phenomena in human osteosarcoma cell lines. As determined by soft agar growth of osteosarcoma cells and xenografted mouse models, ICSBP increased tumorigenicity, which was reversed by ICSBP knock-down or a TGF-ß RI inhibitor. To test whether ICSBP directly regulates the promoter activity of TGF-ß RI, we performed a TGF-ß RI promoter assay, an electro mobility shift assay, and a chromatin immunoprecipitation assay. We observed that TGF-ß RI promoter was activated in ICSBP-overexpressing osteosarcoma cells. Exploiting serial deletions and mutations of the TGF-ß RI promoter, we found a putative ICSBP-binding site at nucleotides -216/-211 (GGXXTC) in the TGF-ß RI promoter. Our data suggest that ICSBP upregulates TGF-ß RI expression by binding to this site, causing ICSBP-mediated tumor progression in osteosarcoma cells. In addition, we found a positive correlation between ICSBP and TGF-ß RI expression in several types of tumors using the cBioportal database. SUMMARY: We demonstrated that interferon consensus sequence-binding protein upregulates transforming growth factor-beta type I receptor (TGF-ß RI) expression by binding to nucleotides -216/-211 (GGXXTC) in the TGF-ß RI promoter, which resulted in increased tumorigenicity and tumor progression in human osteosarcoma cells.


Asunto(s)
Neoplasias Óseas/metabolismo , Regulación Neoplásica de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Proteínas de Neoplasias/metabolismo , Osteosarcoma/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/biosíntesis , Elementos de Respuesta , Regulación hacia Arriba , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Humanos , Factores Reguladores del Interferón/genética , Proteínas de Neoplasias/genética , Osteosarcoma/genética , Osteosarcoma/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética
11.
Biomol Ther (Seoul) ; 27(2): 231-239, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30763986

RESUMEN

Suppressor of Variegation 3-9 Homolog 2 (SUV39H2) methylates the lysine 9 residue of histone H3 and induces heterochromatin formation, resulting in transcriptional repression or silencing of target genes. SUV39H1 and SUV39H2 have a role in embryonic development, and SUV39H1 was shown to suppress cell cycle progression associated with Rb. However, the function of human SUV39H2 has not been extensively studied. We observed that forced expression of SUV39H2 decreased cell proliferation by inducing G1 cell cycle arrest. In addition, SUV39H2 was degraded through the ubiquitin-proteasomal pathway. Using yeast two-hybrid screening to address the degradation mechanism and function of SUV39H2, we identified translationally controlled tumor protein (TCTP) as an SUV39H2-interacting molecule. Mapping of the interacting regions indicated that the N-terminal 60 amino acids (aa) of full-length SUV39H2 and the C-terminus of TCTP (120-172 aa) were critical for binding. The interaction of SUV39H2 and TCTP was further confirmed by co-immunoprecipitation and immunofluorescence staining for colocalization. Moreover, depletion of TCTP by RNAi led to up-regulation of SUV39H2 protein, while TCTP overexpression reduced SUV39H2 protein level. The half-life of SUV39H2 protein was significantly extended upon TCTP depletion. These results clearly indicate that TCTP negatively regulates the expression of SUV39H2 post-translationally. Furthermore, SUV39H2 induced apoptotic cell death in TCTP-knockdown cells. Taken together, we identified SUV39H2, as a novel target protein of TCTP and demonstrated that SUV39H2 regulates cell proliferation of lung cancer cells.

12.
Phytomedicine ; 51: 139-150, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30466611

RESUMEN

BACKGROUND: Afrocyclamin A, an oleanane-type triterpene saponin, was isolated from Androsace umbellata which used as a traditional herbal medicine. PURPOSE: This study aimed to explore the anticancer activity of afrocyclamin A on human prostate cancer cells in vitro as well as in vivo. METHODS: Cytotoxicity, cell cycle distribution, apoptosis, and autophagic cell death were measured following exposure to afrocyclamin A. In vivo antitumor activity of afrocyclamin A was assessed in a xenograft model. The protein levels of p-Akt, p-mTOR, Bax, Bcl-2, caspase-3, and caspase-9 were quantified using western blot analysis. RESULTS: In DU145 cells, afrocyclamin A increased cytotoxicity, caused changes in cell morphology, and induced sub-G0/G1 phase indicating increased apoptosis. Afrocyclamin A robustly induced autophagic cell death as demonstrated by the conversion of LC3B-I to LC3B-II, and the formation of autophagic vacuoles as revealed by western blot analysis and fluorescence staining, respectively. Afrocyclamin A also inhibited the phosphorylation of PI3K, Akt, and mTOR, suggesting their role in afrocyclamin A induced cell death. In addition, afrocyclamin A inhibited cell migration and invasion in concentration and time-dependent manners. In an in vivo xenograft model, afrocyclamin A inhibited the growth of DU145 cells. CONCLUSION: Afrocyclamin A has anticancer activity via the PI3K/Akt/mTOR pathway, which leads to cell death.


Asunto(s)
Autofagia/efectos de los fármacos , Primulaceae/química , Neoplasias de la Próstata/tratamiento farmacológico , Saponinas/farmacología , Transducción de Señal , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fitoquímicos/farmacología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Toxicol Environ Health A ; 81(22): 1173-1185, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30415634

RESUMEN

Cutaneous allergy occurs primarily as a result of using cosmetic, household, and laundry products available on the market that contain fragrances. The aim of this study was to develop a rapid and specific high-performance liquid chromatography with ultraviolet detection (HPLC-UV) method for quantification of 25 fragrance allergens (amyl cinnamyl alcohol, benzyl alcohol, benzyl benzoate, benzyl cinnamate, benzyl salicylate, citronellol, cinnamyl alcohol, citral, coumarin, eugenol, farnesol, geraniol, hydroxycitronellal, HICC (4-(4-hydroxy-4-methylpentyl)-3-cyclohexene-1-carboaldehyde), isoeugenol, isoeugenyl acetate, lilial (butyl phenyl methyl propional), limonene, linalool, methyl 2-octynoate, etc.). In addition, an exposure-based quantitative risk assessment (QRA) was performed to determine safe levels of fragrance ingredients in 107 perfumes. In 76 women's and 31 men's fragrances, 25 allergens were identified at concentrations ranging from undetectable (N.D.) to 8,997.68 mg/kg, and from N.D. to 17,352.34 mg/kg, respectively. An exposure-based sensitization QRA revealed that the ratios of acceptable exposure level (AEL) to consumer exposure level (CEL) of fragrance ingredients were greater than 1, suggesting an absence of skin sensitizing potential. However, the maximum level used in the exposure scenario was determined by the product purpose and application type, and AEL/CEL ratios of lilial, HICC, citral, isoeugenol, and methyl 2-octynoate analyzed in women's perfume were 0.53, 0.67 0.19, 0.13, and 0.57, respectively. As the ratios of AEL:CEL of these fragrance ingredients were below 1, the utilization of these potential skin sensitizers is not considered safe. Our findings indicate that the sensitization risk of allergens with AEL:CEL ratios below 1 detected in fragrances needs to be reduced to the appropriate human safety level for risk management.


Asunto(s)
Alérgenos/análisis , Exposición a Riesgos Ambientales/análisis , Odorantes/análisis , Perfumes/análisis , Dermatitis Alérgica por Contacto , Femenino , Humanos , Masculino , Prevalencia , República de Corea , Medición de Riesgo
14.
Int J Mol Sci ; 19(9)2018 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-30217020

RESUMEN

We previously discovered a novel sirtuin (SIRT) inhibitor, MHY2256, that exerts anticancer activity through p53 acetylation in MCF-7 human breast cancer cells. We investigated the anticancer activity of MHY2256 against hormone-related cancer, an endometrial cancer with a poor prognosis. The IC50 values of MHY2256 were shown to be much lower than those of salermide, a well-known SIRT inhibitor. Furthermore, MHY2256 significantly reduced the protein expression and activities of SIRT1, 2, and 3, with similar effects to salermide. Particularly, MHY2256 markedly inhibited tumor growth in a tumor xenograft mouse model of Ishikawa cancer cells. During the experimental period, there was no significant change in the body weight of mice treated with MHY2256. A detailed analysis of the sensitization mechanisms of Ishikawa cells revealed that late apoptosis was largely increased by MHY2256. Additionally, MHY2256 increased G1 arrest and reduced the number of cell cyclic-related proteins, suggesting that apoptosis by MHY2256 was achieved by cellular arrest. Particularly, p21 was greatly increased by MHY225656, suggesting that cell cycle arrest by p21 is a major factor in MHY2256 sensitization in Ishikawa cells. We also detected a significant increase in acetylated p53, a target protein of SIRT1, in Ishikawa cells after MHY2256 treatment. In a mouse xenograft model, MHY2256 significantly reduced tumor growth and weight without apparent side effects. These results suggest that MHY2256 exerts its anticancer activity through p53 acetylation in endometrial cancer and can be used for targeting hormone-related cancers.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias Endometriales/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Animales , Western Blotting , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
15.
J Toxicol Environ Health A ; 81(17): 830-843, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30020862

RESUMEN

Potential biomarkers of skin sensitization in RAW264.7 mouse macrophages were investigated as alternatives to animal experiments and risk assessment. The concentrations that resulted in a cell viability of 90% (CV90) and 75% (CV75) were calculated by using a water-soluble tetrazolium salt (WST)-1 assay and used to analyze the skin sensitization potency of 23 experimental materials under equivalent treatment conditions. In addition, the expression of interleukin (IL)-1α, IL-1ß, IL-31, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS), prostaglandin E2 (PGE2), and cyclooxygenase-2 (COX-2) was analyzed utilizing Western blotting. In the cell viability analysis, skin sensitizers were generally more cytotoxic and exhibited increased skin sensitization potency. However, nonsensitizers did not show any marked cytotoxic tendency. Biomarker analysis demonstrated that IL-1α, IL-1ß, and the combination of IL-1α and IL-1ß (IL-1α + IL-1ß) predicted reliably skin sensitization potential (1) sensitivities of 94.4%, 83.3%, and 83.3%, specificities of 100%, 100%, and 100%, and (2) accuracies of 95.7%, 87%, and 87%, respectively. These observations correlated most reliably as indicators for skin sensitization potency. Data suggest that IL-1α and IL-1ß may serve as potential biomarkers for skin sensitization and provide an alternative method to animal experiments for prediction of skin sensitization potency and risk assessment.


Asunto(s)
Alternativas a las Pruebas en Animales/métodos , Dermatitis Alérgica por Contacto/fisiopatología , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Piel/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Dermatitis Alérgica por Contacto/etiología , Ratones , Células RAW 264.7 , Medición de Riesgo/métodos
16.
BMB Rep ; 48(2): 115-20, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25027596

RESUMEN

Tight junction protein 1 (TJP1), a component of tight junction, has been reported to play a role in protein networks as an adaptor protein, and TJP1 expression is altered during tumor development. Here, we found that TJP1 expression was increased at the RNA and protein levels in TGF-ß-stimulated lung cancer cells, A549. SB431542, a type-I TGF-ß receptor inhibitor, as well as SB203580, a p38 kinase inhibitor, significantly abrogated the effect of TGF-ß on TJP1 expression. Diphenyleneiodonium, an NADPH oxidase inhibitor, also attenuated TJP1 expression in response to TGF-ß in lung cancer cells. When TJP1 expression was reduced by shRNA lentiviral particles in A549 cells (A549-sh TJP1), wound healing was much lower than in cells infected with control viral particles. Taken together, these data suggest that TGF-ß enhances TJP1 expression, which may play a role beyond structural support in tight junctions during cancer development.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteína de la Zonula Occludens-1/metabolismo , Benzamidas/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dioxoles/farmacología , Humanos , Imidazoles/farmacología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Compuestos Onio/farmacología , Piridinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína de la Zonula Occludens-1/antagonistas & inhibidores , Proteína de la Zonula Occludens-1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Cell Signal ; 26(11): 2562-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25086206

RESUMEN

Wnt inhibitory factor-1 (WIF1) is a conserved lipid-binding protein that interrupts Wnt ligands by interacting with their Frizzled receptors. Thus, they may suppress the activation of the Wnt/ß-catenin triggered signaling cascade. Recently, we found that WIF1 can effectively co-regulate pro-apoptotic activity through the combination with Dickkopf-1 (DKK1). The tumor suppressor p53 protein expression was remarkably increased in the WIF1- and DKK1-transfected cells, along with p21. In contrast, expressions of the anti-apoptotic proteins, c-Myc and Bcl-2, were noticeably reduced. In addition, WIF1 and/or DKK1 significantly activated the transcription of p21 and p53, whereas c-Myc and Bcl-2 activities were remarkably reduced. The tumor suppressor WIF1 was also found to be capable of suppressing tumor growth through the inhibition of tumor angiogenesis in the cellular biological/physiological condition through the targeting of the PI3K/Akt/mTOR signaling pathway, while also being recognized as a Wnt antagonist factor in the Wnt cascade. Consistently, WIF1 conspicuously decreased the VEGF-induced phosphorylation of the PI3K/Akt signaling cascade components, including PDK1, mTOR, TSC-2, GSK-3ß, and the p70S6K protein. Collectively, our results indicate for the first time that the tumor suppressor WIF1 is involved in angiogenesis and supplies a possible molecular target for the treatment of distinct malignant cancers, as well as several other associated diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Represoras/metabolismo , Vía de Señalización Wnt , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
18.
Oncotarget ; 5(15): 6540-51, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25153728

RESUMEN

Recently, we found that sMEK1 effectively regulates pro-apoptotic activity when combined with a traditional chemotherapeutic drug. Therefore, combinational therapeutic strategies targeting critical molecular and cellular mechanisms are urgently required. In this present work, we evaluated whether sMEK1 enhanced the pro-apoptotic activity of chemotherapeutic drugs in ovarian carcinoma cells. Combined with a chemotherapeutic drug, sMEK1 showed an additive effect on the suppression of ovarian cancer cell growth by inducing cell cycle arrest and apoptosis and regulating related gene expression levels or protein activities. In addition, the phosphoinositide-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway was strongly inhibited by the combined treatment, showing de-repression of the tuberous sclerosis complex (TSC) and suppression of ras homolog enriched in the brain (Rheb) and mTOR and raptor in aggressive ovarian carcinoma cells and mouse xenograft models. Treatment with sMEK1 and paclitaxel reduced phosphorylation of ribosomal S6 kinase (S6K) and 4E-binding protein (4E-BP), two critical downstream targets of the mTOR-signaling pathway. Furthermore, both sMEK1 and paclitaxel significantly inhibited the expression of signaling components downstream of S6K/4E-BP, such as hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF), both in vitro and in vivo. Therefore, our data suggest that the combination of sMEK1 and paclitaxel is a promising and effective targeted therapy for chemotherapy-resistant or recurrent ovarian cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Epitelial de Ovario , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Complejos Multiproteicos/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Paclitaxel/administración & dosificación , Fosfoproteínas Fosfatasas/administración & dosificación , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-24749480

RESUMEN

Endocrine-disrupting chemicals (EDC), including phthalates, bisphenol A (BPA), phytoestrogens such as genistein and daidzein, dichlorodiphenyltrichloroethane (DDT), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are associated with a variety of adverse health effects in organisms or progeny by altering the endocrine system. Environmental estrogens, including BPA, phthalates, and phytoestrogens, are the most extensively studied and are considered to mimic the actions of endogenous estrogen, 17ß-estradiol (E2). Diverse modes of action of estrogen and estrogen receptors (ERα and ERß) have been described, but the mode of action of estrogenic EDC is postulated to be more complex and needs to be more clearly elucidated. This review examines the adverse effects of estrogenic EDC on male or female reproductive systems and molecular mechanisms underlying EDC effects that modulate ER-mediated signaling. Mechanisms of action for estrogenic EDC may involve both ER-dependent and ER-independent pathways. Recent findings from systems toxicology of examining estrogenic EDC are also discussed.


Asunto(s)
Sistema Endocrino/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Estrógenos/toxicidad , Genitales/efectos de los fármacos , Animales , Exposición a Riesgos Ambientales , Femenino , Humanos , Masculino
20.
Artículo en Inglés | MEDLINE | ID: mdl-23573938

RESUMEN

d-Limonene, a major constituent of citrus oils, is a monoterpene widely used as a flavor/fragrance additive in cosmetics, foods, and industrial solvents as it possesses a pleasant lemon-like odor. d-Limonene has been designated as a chemical with low toxicity based upon lethal dose (LD50) and repeated-dose toxicity studies when administered orally to animals. However, skin irritation or sensitizing potential was reported following widespread use of this agent in various consumer products. In experimental animals and humans, oxidation products or metabolites of d-limonene were shown to act as skin irritants. Carcinogenic effects have also been observed in male rats, but the mode of action (MOA) is considered irrelevant for humans as the protein α(2u)-globulin responsible for this effect in rodents is absent in humans. Thus, the liver was identified as a critical target organ following oral administration of d-limonene. Other than the adverse dermal effects noted in humans, other notable toxic effects of d-limonene have not been reported. The reference dose (RfD), the no-observed-adverse-effect level (NOAEL), and the systemic exposure dose (SED) were determined and found to be 2.5 mg/kg/d, 250 mg/kg//d, and 1.48 mg/kg/d, respectively. Consequently, the margin of exposure (MOE = NOAEL/SED) of 169 was derived based upon the data, and the hazard index (HI = SED/RfD) for d-limonene is 0.592. Taking into consideration conservative estimation, d-limonene appears to exert no serious risk for human exposure. Based on adverse effects and risk assessments, d-limonene may be regarded as a safe ingredient. However, the potential occurrence of skin irritation necessitates regulation of this chemical as an ingredient in cosmetics. In conclusion, the use of d-limonene in cosmetics is safe under the current regulatory guidelines for cosmetics.


Asunto(s)
Ciclohexenos/toxicidad , Terpenos/toxicidad , Animales , Carcinógenos/toxicidad , Relación Dosis-Respuesta a Droga , Aromatizantes/toxicidad , Humanos , Limoneno , Masculino , Concentración Máxima Admisible , Ratones , Ratas , Medición de Riesgo/métodos , Medición de Riesgo/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...