Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Yonsei Med J ; 57(3): 588-98, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26996556

RESUMEN

PURPOSE: Tolfenamic acid (TA), a non-steroidal anti-inflammatory drug, is known to exhibit antitumor effects in various cancers apart from nasopharyngeal cancer (NPC). NPC exhibits high invasiveness, as well as metastatic potential, and patients continue to suffer from residual, recurrent, or metastatic disease even after chemoradiation therapy. Therefore, new treatment strategies are needed for NPC. In this study, we investigated the efficacy and molecular mechanisms of TA in NPC treatment. MATERIALS AND METHODS: TA-induced cell death was detected by cell viability assay in the NPC cell lines, HNE1 and HONE1. Wound healing assay, invasion assay, and Western blot analysis were used to evaluate the antitumor effects of TA in NPC cell lines. RESULTS: Treatment with TA suppressed the migration and invasion of HNE1 and HONE1 cells. Hepatocyte growth factor enhanced the proliferation, migration, and invasion abilities of NPC cells. This enhancement was successfully inhibited by TA treatment. Treatment with TA increased phosphorylation of p38, and the inhibition of p38 with SB203580 reversed the cytotoxic, anti-invasive, and anti-migratory effects of TA treatment in NPC cell lines. Moreover, inhibition of p38 also reversed the decrease in expression of Slug that was induced by TA treatment. CONCLUSION: In conclusion, the activation of p38 plays a role in mediating TA-induced cytotoxicity and inhibition of invasion and migration via down-regulation of Slug.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Invasividad Neoplásica/prevención & control , ortoaminobenzoatos/farmacología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Carcinoma , Línea Celular Tumoral , Regulación hacia Abajo , Gastrópodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Imidazoles , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Fosforilación/efectos de los fármacos , Piridinas , ortoaminobenzoatos/uso terapéutico
2.
Cancer Res Treat ; 48(2): 738-52, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26511816

RESUMEN

PURPOSE: The purpose of this study is to determine whether luminacin, a marine microbial extract from the Streptomyces species, has anti-tumor effects on head and neck squamous cell carcinoma (HNSCC) cell lines via autophagic cell death. MATERIALS AND METHODS: Inhibition of cell survival and increased cell death was measured using cell viability, colony forming, and apoptosis assays. Migration and invasion abilities of head and cancer cells were evaluated using wound healing, scattering, and invasion assays. Changes in the signal pathway related to autophagic cell death were investigated. Drug toxicity of luminacin was examined in in vitro HaCaT cells and an in vivo zebrafish model. RESULTS: Luminacin showed potent cytotoxicity in HNSCC cells in cell viability, colony forming, and fluorescence-activated cell sorting analysis. In vitro migration and invasion of HNSCC cells were attenuated by luminacin treatment. Combined with Beclin-1 and LC3B, Luminacin induced autophagic cell death in head and neck cancer cells. In addition, in a zebrafish model and human keratinocyte cell line used for toxicity testing, luminacin treatment with a cytotoxic concentration to HNSCC cells did not cause toxicity. CONCLUSION: Taken together, these results demonstrate that luminacin induces the inhibition of growth and cancer progression via autophagic cell death in HNSCC cell lines, indicating a possible alternative chemotherapeutic approach for treatment of HNSCC.


Asunto(s)
Autofagia/efectos de los fármacos , Benzaldehídos/farmacología , Benzaldehídos/uso terapéutico , Carcinoma de Células Escamosas/patología , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico , Neoplasias de Cabeza y Cuello/patología , Compuestos de Espiro/farmacología , Compuestos de Espiro/uso terapéutico , Animales , Benzaldehídos/efectos adversos , Carcinoma de Células Escamosas/tratamiento farmacológico , Extractos Celulares/efectos adversos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Progresión de la Enfermedad , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Invasividad Neoplásica/prevención & control , Compuestos de Espiro/efectos adversos , Pez Cebra/embriología
3.
Phytomedicine ; 22(6): 679-88, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26055133

RESUMEN

BACKGROUND: Oral mucositis is a common adverse effect of antineoplastic chemotherapy limiting sufficient dose of chemoregimen. Numerous attempts to mitigate chemotherapy-induced oral mucositis have failed to identify an appropriate treatment. HYPOTHESIS: We hypothesize that Artemisia asiatica (Pamp.) Nakai ex Kitam ethanol extract (Aa-EE) would mitigate cisplatin-induced cytotoxicity to oral mucosal epithelial cells. STUDY DESIGN: In vitro experimental study. METHODS: Cell viability and wound healing assay were performed. Apoptosis, mitochondrial membrane potential (MMP) change, and changes in apoptosis-related signaling were demonstrated in human primary keratinocyte (HaCaT). RESULTS: Cisplatin inhibited HaCaT cell proliferation and migration. Aa-EE protected against these effects. Cisplatin treatment of HaCaT cells caused apoptosis and changes in MMP. Aa-EE inhibited cisplatin-induced apoptosis, and stabilized the cisplatin-induced loss of MMP. Western blots revealed that Aa-EE reduced the expression of cytochrome c and cleaved caspase-3 and inhibited nuclear translocation of nuclear factor-kappa B (NF-κB), compared with the levels observed after cisplatin treatment, whereas Bcl-2 expression was increased by Aa-EE. CONCLUSION: Collectively, our results suggest that Aa-EE protects HaCaT cells by inhibiting cisplatin-induced mitochondrial damage associated with Bcl-2 activity and by inhibiting nuclear translocation of NF-κB.


Asunto(s)
Artemisia/química , Queratinocitos/efectos de los fármacos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Cisplatino/efectos adversos , Flavonoides/farmacología , Humanos , Potencial de la Membrana Mitocondrial , Componentes Aéreos de las Plantas/química , Transducción de Señal
4.
Radiat Oncol J ; 33(4): 328-36, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26756033

RESUMEN

PURPOSE: Past studies have reported that S-allylcysteine (SAC) inhibits the migration and invasion of cancer cells through the restoration of E-cadherin, the reduction of matrix metalloproteinase (MMP) and Slug protein expression, and inhibition of the production of reactive oxygen species (ROS). Furthermore, evidence is emerging that shows that ROS induced by radiation could increase Met activation. Following on these reports of SAC and Met, we investigated whether SAC could suppress Met activation. MATERIALS AND METHODS: Wound healing, invasion, 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium (MTT), soft agar colony forming, western blotting, and gelatin zymography assays were performed in the human nasopharyngeal cancer cell lines HNE1 and HONE1 treated with SAC (0, 10, 20, or 40 mM) and hepatocyte growth factor (HGF). RESULTS: This study showed that SAC could suppress the migration and invasion of HNE1 and HONE1 cell lines by inhibiting p-Met. An increase of migration and invasion induced by HGF and its decrease in a dose dependent manner by SAC in wound healing and invasion assays was observed. The reduction of p-Met by SAC was positively correlated with p-focal adhesion kinase (p-FAK) and p-extracellular related kinase (p-ERK in both cell lines). SAC reduced Slug, MMP2, and MMP9 involved in migration and invasion with the inhibition of Met-FAK signaling. CONCLUSION: These results suggest that SAC inhibited not only Met activation but also the downstream FAK, Slug, and MMP expression. Finally, SAC may be a potent anticancer compound for nasopharyngeal cancer treated with radiotherapy.

5.
Lab Anim Res ; 30(2): 45-53, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24999358

RESUMEN

Pig pancreas may be a therapeutic resource for human diabetic patients. However, this potential is hindered by a lack of knowledge of the molecular events of pig pancreas development. In this study, the embryonic day 60, neonate and 6-month protein profiles of pig pancreas were ascertained at using two-dimensional gel electrophoresis and matrix assisted laser desorption/ionization-time of flight mass spectrometry. Twenty four proteins were differentially expressed during pig pancreas development. Among them, 12 spots increased and 7 spots decreased according to development. The expression of 5 protein were highest at birth. Expression of digestive enzymes including trypsin, pancreatic triacylglycerol lipase and pancreatic alpha-amylase was elevated in adults, whereas chymotrypsins were highly expressed in neonates. Proteins that were abundantly expressed during gestation were alpha-1-antitrypsin, alpha-fetoprotein and transferrins. Taken together, we found out that several proteins were significantly up- or down- regulated from pig pancreas based on developmental stage. This study will provide basis for understanding development of pig pancreas.

6.
Yonsei Med J ; 55(4): 886-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24954315

RESUMEN

PURPOSE: Acute side effects of radiation such as oral mucositis are observed in most patients. Although several potential radioprotective agents have been proposed, no effective agent has yet been identified. In this study, we investigated the effectiveness of synthetic compound 3-amino-3-(4-fluoro-phenyl)-1H-quinoline-2,4-dione (KR22332) as a radioprotective agent. MATERIALS AND METHODS: Cell viability, apoptosis, the generation of reactive oxygen species (ROS), mitochondrial membrane potential changes, and changes in apoptosis-related signaling were examined in human keratinocyte (HaCaT). RESULTS: KR22332 inhibited irradiation-induced apoptosis and intracellular ROS generation, and it markedly attenuated the changes in mitochondrial membrane potential in primary human keratinocytes. Moreover, KR22332 significantly reduced the protein expression levels of ataxia telangiectasia mutated protein, p53, and tumor necrosis factor (TNF)-α compared to significant increases observed after radiation treatment. CONCLUSION: KR22332 significantly inhibited radiation-induced apoptosis in human keratinocytes in vitro, indicating that it might be a safe and effective treatment for the prevention of radiation-induced mucositis.


Asunto(s)
Protectores contra Radiación/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Humanos , Queratinocitos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Protectores contra Radiación/química , Especies Reactivas de Oxígeno/metabolismo
7.
Neurotoxicology ; 40: 111-22, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24374476

RESUMEN

Radiation is a widely used treatment for head and neck cancers, and one of its most severe side effects is ototoxicity. Radiation-induced ototoxicity has been demonstrated to be linked to the increased production of ROS and MAPK. We intended to investigate the effect of p38 inhibition on radiation-induced ototoxicity in cochlea-derived HEI-OC1 cells and in a zebrafish model. The otoprotective effect of p38 inhibition against radiation was tested in vitro in the organ of Corti-derived cell line, HEI-OC1, and in vivo in a zebrafish model. Radiation-induced apoptosis, mitochondrial dysfunction, and an increase of intracellular NO generation were demonstrated in HEI-OC1 cells. The p38-specific inhibitor, SB203580, ameliorated radiation-induced apoptosis and mitochondrial injury in HEI-OC1 cells. p38 inhibition reduced radiation-induced activation of JNK, p38, cytochrome c, and cleavage of caspase-3 and PARP in HEI-OC1 cells. Scanning electron micrography showed that SB203580 prevented radiation-induced destruction of kinocilium and stereocilia in zebrafish neuromasts. The results of this study suggest that p38 plays an important role in mediating radiation-induced ototoxicity and inhibition of p38 could be a plausible option for preventing radiation ototoxicity.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Imidazoles/uso terapéutico , Órgano Espiral/efectos de la radiación , Piridinas/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Muerte Celular , Línea Celular , Células Ciliadas Auditivas/efectos de los fármacos , Células Ciliadas Auditivas/efectos de la radiación , Células Ciliadas Auditivas/ultraestructura , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Órgano Espiral/efectos de los fármacos , Órgano Espiral/patología , Fotones , Pez Cebra
8.
J Appl Toxicol ; 34(2): 191-204, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23297007

RESUMEN

Cisplatin [cis-diammine-dichloroplatinum (II)] is a widely used chemotherapeutic agent, and one of its most severe side effects is ototoxicity. In the course of developing a new protective agent against cisplatin-induced ototoxicity, we have been interested in a novel synthetic compound, 3-Amino-3-(4-fluoro-phenyl)-1H-quinoline-2,4-dione (KR-22335). We evaluated the effectiveness of KR-22335 as an otoprotective agent against cisplatin-induced toxicity. The otoprotective effect of KR-22335 against cisplatin was tested in vitro in cochlear organs of Corti-derived cell lines, HEI-OC1, and in vivo in a zebrafish (Danio rerio) model. Cisplatin-induced apoptosis, cell cycle arrest and an increase in intracellular reactive oxygen species (ROS) generation were demonstrated in HEI-OC1 cells. KR-22335 inhibited cisplatin-induced apoptosis and mitochondrial injury in HEI-OC1 cells. KR-22335 inhibited cisplatin-induced activation of JNK, p-38, caspase-3 and PARP in HEI-OC1 cells. Scanning and transmission electron micrographs showed that KR-22335 prevented cisplatin-induced destruction of kinocilium and stereocilia in zebrafish neuromasts. Tissue TUNEL of neuromasts in zebrafish demonstrated that KR-22335 blocked cisplatin-induced TUNEL positive hair cells in neuromasts. The results of this study suggest that KR-22335 may prevent ototoxicity caused by the administration of cisplatin through the inhibition of mitochondrial dysfunction and suppression of ROS generation. KR-22335 may be considered as a potential candidate for protective agents against cisplatin-induced ototoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Cisplatino/efectos adversos , Células Ciliadas Auditivas/efectos de los fármacos , Sustancias Protectoras/farmacología , Quinolonas/farmacología , Animales , Línea Celular Tumoral , Células Ciliadas Auditivas/citología , Humanos , Etiquetado Corte-Fin in Situ , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra
9.
J Radiat Res ; 55(2): 245-56, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24078877

RESUMEN

Radiation-induced oral mucositis is a dose-limiting toxic side effect for patients with head and neck cancer. Numerous attempts at improving radiation-induced oral mucositis have not produced a qualified treatment. Ginseng polysaccharide has multiple immunoprotective effects. Our aim was to investigate the effectiveness of Korean red ginseng (KRG) on radiation-induced damage in the human keratinocyte cell line HaCaT and in an in vivo zebrafish model. Radiation inhibited HaCaT cell proliferation and migration in a cell viability assay and wound healing assay, respectively. KRG protected against these effects. KRG attenuated the radiation-induced embryotoxicity in the zebrafish model. Irradiation of HaCaT cells caused apoptosis and changes in mitochondrial membrane potential (MMP). KRG inhibited the radiation-induced apoptosis and intracellular generation of reactive oxygen species (ROS), and stabilized the radiation-induced loss of MMP. Western blots revealed KRG-mediated reduced expression of ataxia telangiectasia mutated protein (ATM), p53, c-Jun N-terminal kinase (JNK), p38 and cleaved caspase-3, compared with their significant increase after radiation treatment. The collective results suggest that KRG protects HaCaT cells by blocking ROS generation, inhibiting changes in MMP, and inhibiting the caspase, ATM, p38 and JNK pathways.


Asunto(s)
Queratinocitos/fisiología , Queratinocitos/efectos de la radiación , Panax/química , Extractos Vegetales/administración & dosificación , Tolerancia a Radiación/fisiología , Protectores contra Radiación/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Queratinocitos/citología , Corea (Geográfico) , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Estrés Oxidativo/efectos de la radiación , Tolerancia a Radiación/efectos de los fármacos
10.
Laryngoscope ; 122(10): 2256-64, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22815072

RESUMEN

OBJECTIVES/HYPOTHESIS: Tumor necrosis factor-alpha (TNF-α) is released in a variety of pathological states in the inner ear. Inducible nitric oxide synthase (iNOS) can be induced by cytokines and other inflammatory factors, and is generally thought to be associated with inflammation and other pathological processes in the cochlea. The purpose of the present study was to reveal that TNF-α could induce apoptosis in the auditory cell line and to investigate the role of nitric oxide (NO) in TNF-α-induced auditory cell death. STUDY DESIGN: Experimental study. METHODS: UB-OC1 cells and zebrafish were exposed to TNF-α. Flow cytometry, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assay, assay of mitochondrial membrane potential (MMP), and electron microscopy were used to show that TNF-α could induce apoptosis. Western blot was used to measure iNOS expression and mitogen-activated protein kinase pathway. RESULTS: Flow cytometric analysis, TUNEL assay, MMP, and electron microscopy all demonstrated that TNF-α could induce apoptosis in UB-OC1 cells. TNF-α significantly increased NO generation and iNOS expression. Pretreatment with iNOS blocker NG-methyl-L-arginine (NMA) attenuated TNF-α-induced cell death and caspase-3 activation. Also, TNF-α treatment increased p-p38 and p-ERK, and pretreatment of NMA reduced this increased expression of p-p38 and p-ERK. CONCLUSIONS: TNF-α can induce apoptosis in the auditory cell line, and NO production in response to TNF-α is essential for apoptosis.


Asunto(s)
Apoptosis/fisiología , Células Ciliadas Auditivas Internas/metabolismo , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Supervivencia Celular , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/ultraestructura , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Pez Cebra
11.
PLoS One ; 7(4): e34988, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536345

RESUMEN

Nonsteroidal anti-inflammatory drug-activated gene-1 (NAG-1) is induced by nonsteroidal anti-inflammatory drugs and possesses proapoptotic and antitumorigenic activities. Although tolfenamic acid (TA) induces apoptosis in head and neck cancer cells, the relationship between NAG-1 and TA has not been determined. This study investigated the induction of apoptosis in head and neck cancer cells treated by TA and the role of NAG-1 expression in this induction. TA reduced head and neck cancer cell viability in a dose-dependent manner and induced apoptosis. The induced apoptosis was coincident with the expression of NAG-1. Overexpression of NAG-1 enhanced the apoptotic effect of TA, whereas suppression of NAG-1 expression by small interfering RNA attenuated TA-induced apoptosis. TA significantly inhibited tumor formation as assessed by xenograft models, and this result accompanied the induction of apoptotic cells and NAG-1 expression in tumor tissue samples. Taken together, these results demonstrate that TA induces apoptosis via NAG-1 expression in head and neck squamous cell carcinoma, providing an additional mechanistic explanation for the apoptotic activity of TA.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Factor 15 de Diferenciación de Crecimiento/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , ortoaminobenzoatos/farmacología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Expresión Génica , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Activación Transcripcional/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , ortoaminobenzoatos/uso terapéutico
12.
Cancer Lett ; 322(1): 45-57, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22353688

RESUMEN

The proapoptotic death receptor 4 (DR4), along with DR5, is currently regarded as a promising target for development of agonistic anti-cancer agents due to its tumor-selective apoptosis-inducing ability with no significant cytotoxicity to normal cells. In this study, we examine susceptibility of various head and neck cancer (HNC) cells and mechanism of cell death to an anti-DR4 agonistic monoclonal antibody (mAb), AY4. AY4 as a single agent induced caspase-dependent apoptotic cell death of KB and HN9, but not in SNU899 and FaDu cell lines. AY4 treatment resulted in accumulation of intracellular reactive oxygen species (ROS) generated from mitochondria in AY4-sensitive cells. Blockade of ROS production by N-acetyl-l-cysteine (NAC) resulted in protection of AY4-sensitive cells against AY4-induced apoptosis. ROS generation induced by AY4 treatment triggered down-regulation of anti-apoptotic molecules of Bcl-xL and X-linked inhibitor of apoptosis (XIAP) without affecting the expression levels of DR4, Mcl-1, and survivin. AY4 also inhibited growth of pre-established HN9 tumors in a nude mouse xenograft model and did not show noticeable cytotoxicity in a zebrafish model. Our results provide further insight into the mechanism of DR4-mediated cell death and potential use of AY4 mAb as an anti-cancer therapeutic agent in AY4-sensitive HNC types.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Acetilcisteína/farmacología , Animales , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/análisis , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/análisis , Proteína Inhibidora de la Apoptosis Ligada a X/antagonistas & inhibidores , Pez Cebra , Proteína bcl-X/análisis , Proteína bcl-X/antagonistas & inhibidores
13.
Yonsei Med J ; 52(2): 293-300, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21319349

RESUMEN

PURPOSE: The risk of invasive fungal infection is greater for allogeneic hematopoietic stem cell transplantation (HSCT) than for autologous transplantation. Therefore, many transplantation centers use antifungal prophylaxis for allogeneic HSCT, however, there exists no standard guidelines or consensus regarding autologous HSCT. MATERIALS AND METHODS: A prospective double-blind randomized study was conducted in autologous HSCT recipients who were divided into prophylaxis and empirical treatment groups, and we investigated the efficacy of itraconazole prophylaxis in pediatric autologous HSCT. RESULTS: Total 87 autologous HSCT episodes in 55 children with high-risk solid tumors were studied. No invasive fungal infections occurred in either group. However, patients in the prophylaxis group had a significantly shorter duration of fever (p < 0.05) and received antibacterial treatment of shorter duration (p < 0.05) with fewer numbers of antibiotics (p < 0.05 for the use of second line antibiotics) than those in the empirical group. No significant additional adverse events were found with itraconazole prophylaxis. CONCLUSION: Although beneficial effects such as a shorter duration of fever and reduced need for antibiotic use were observed in the prophylaxis group, the results were not sufficient to draw a definite recommendation about the routine use of antifungal prophylaxis in pediatric autologous HSCT recipients with high-risk solid tumors (Trial registration: NCT00336531).


Asunto(s)
Antifúngicos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Itraconazol/uso terapéutico , Micosis/prevención & control , Neoplasias/cirugía , Antibacterianos/uso terapéutico , Preescolar , Análisis Costo-Beneficio , Método Doble Ciego , Humanos , Estudios Prospectivos , Factores de Riesgo , Trasplante Autólogo , Resultado del Tratamiento
14.
J Nutr Biochem ; 22(11): 1074-83, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21292466

RESUMEN

Hepatocyte growth factor (HGF) and c-Met have recently attracted a great deal of attention as prognostic indicators of patient outcome, and they are important in the control of tumor growth and invasion. Epigallocatechin-3-gallate (EGCG) has been shown to modulate multiple signal pathways in a manner that controls the unwanted proliferation and invasion of cells, thereby imparting cancer chemopreventive and therapeutic effects. In this study, we investigated the effects of EGCG in inhibiting HGF-induced tumor growth and invasion of oral cancer in vitro and in vivo. We examined the effects of EGCG on HGF-induced cell proliferation, migration, invasion, induction of apoptosis and modulation of HGF/c-Met signaling pathway in the KB oral cancer cell line. We investigated the antitumor effect and inhibition of c-Met expression by EGCG in a syngeneic mouse model (C3H/HeJ mice, SCC VII/SF cell line). HGF promoted cell proliferation, migration, invasion and induction of MMP (matrix metalloproteinase)-2 and MMP-9 in KB cells. EGCG significantly inhibited HGF-induced phosphorylation of Met and cell growth, invasion and expression of MMP-2 and MMP-9. EGCG blocked HGF-induced phosphorylation of c-Met and that of the downstream kinases AKT and ERK, and inhibition of p-AKT and p-ERK by EGCG was associated with marked increases in the phosphorylation of p38, JNK, cleaved caspase-3 and poly-ADP-ribose polymerase. In C3H/HeJ syngeneic mice, as an in vivo model, tumor growth was suppressed and apoptosis was increased by EGCG. Our results suggest that EGCG may be a potential therapeutic agent to inhibit HGF-induced tumor growth and invasion in oral cancer.


Asunto(s)
Catequina/análogos & derivados , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Animales , Catequina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimioprevención , Progresión de la Enfermedad , Femenino , Humanos , Células KB , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Neoplasias de la Boca/patología , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-met/biosíntesis , Transducción de Señal/efectos de los fármacos ,
15.
Head Neck ; 32(12): 1655-64, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20848408

RESUMEN

BACKGROUND: Hepatocyte growth factor (HGF) can promote proliferation, invasion, and angiogenesis in various tumor cells. However, the prognosis according to expression of HGF/c-Met has not been reported in patients with squamous cell carcinoma of the oral tongue (SCCOT). METHODS: Tumors from 61 patients with SCCOT were evaluated for the expression of HGF and c-Met by immunohistochemistry. For Western blot, we used fresh normal and cancer tissues from 3 patients with SCCOT. RESULTS: The positive rates of HGF and c-Met immunostaining in SCCOT were 57.3% and 54.1%, respectively. The c-Met staining was significantly correlated with lymph node metastasis (p = .005), tumor classification (p = .004), and recurrence (p = .018). Survival was significantly affected in patients with positive c-Met expression (p = .003). HGF and c-Met were strongly expressed in cancer tissues on Western blot. CONCLUSION: The c-Met expression may play an important role in the progression and the survival outcome of patients with SCCOT.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Neoplasias de la Lengua/metabolismo , Adulto , Anciano , Western Blotting , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Supervivencia sin Enfermedad , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Análisis de Supervivencia , Neoplasias de la Lengua/mortalidad , Neoplasias de la Lengua/patología , Adulto Joven
16.
Arch Dermatol Res ; 302(4): 301-6, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20033192

RESUMEN

Development of vitiligo-like hypopigmentary lesions associated with topical imiquimod has been reported. We hypothesized that mode of action of imiquimod in melanocytes may include triggering of apoptosis resulted in loss of cells, which may be a possible mechanism of imiquimod-induced hypopigmentary lesions. Therefore, we investigated whether imiquimod induces apoptosis of human melanocytes and also whether it modulates expression of apoptosis-related molecules in human melanocytes. Imiquimod treatment induced apoptosis of melanocytes, which was observed by TUNEL assay and Hoechst 33258 staining. Imiquimod-induced apoptosis was further shown by measuring mitochondrial membrane potential in melanocytes. The apoptotic activity of imiquimod was associated with caspase-3, Bcl-2 and mitogen-activated protein kinase expression in melanocytes. These results indicated that imiquimod induces apoptosis of melanocytes. These findings may provide a clue to understand pathogenesis of imiquimod-induced vitiligo-like hypopigmentary lesions.


Asunto(s)
Aminoquinolinas/efectos adversos , Melanocitos/efectos de los fármacos , Vitíligo/inducido químicamente , Adyuvantes Inmunológicos/efectos adversos , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Células Cultivadas , Humanos , Hipopigmentación , Imiquimod , Etiquetado Corte-Fin in Situ , Melanocitos/metabolismo , Melanocitos/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Vitíligo/metabolismo , Vitíligo/patología , Vitíligo/fisiopatología
17.
Clin Exp Otorhinolaryngol ; 3(4): 217-25, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21217964

RESUMEN

OBJECTIVES: To determine whether a novel marine micro-organism with anticancer properties, H31, the metabolic product of Bacillus SW31, has anti-tumor effects on head and neck cancer, and potential for apoptotic-enhancing anti-cancer treatment of affected patients. METHODS: The cell viability and apoptosis assays were performed. Changes in the signal pathway related to apoptosis were investigated. Then, the therapeutic effects of H31 were explored in mouse xenograft model and drug toxicity of H31 was examined in zebrafish model. RESULTS: We identified the anticancer activity of H31, a novel metabolic product of Bacillus SW31. Bacillus SW31, a new marine micro-organism, has 70% homology with Bacillus firmus and contains potent cytotoxic bioactivity in head and neck cancer cells using MTT assay. Combined with c-JUN, p53, cytochrome C, and caspase-3, H31 induced apoptosis of KB cells, a head and neck cancer cell line. In a separate in vivo model, tumor growth in C3H/HeJ syngeneic mice was suppressed by H31. In addition, in a zebrafish model used for toxicity testing, a considerable dose of H31 did not result in embryo or neurotoxicity. CONCLUSION: Growth inhibition and apoptosis were achieved both in vitro and in vivo in head and neck cancer cells after exposure to H31, a metabolite from the marine Bacillus species, without any significant toxicity effects even at considerable H31 dose concentrations.

18.
Brain Res ; 1263: 134-42, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19368835

RESUMEN

Chronological changes of tissue-type transglutaminase (tTG) were observed in the hippocampal CA1 region after transient forebrain ischemia in gerbils. In the sham-operated group, tTG immunoreactivity was weakly detected in blood vessels which were immunostained with platelet endothelial cell adhesion molecule-1 (PECAM-1), and tTG immunoreactivity in blood vessels was highest 5 days after ischemia/reperfusion. In addition, tTG immunoreaction was expressed in microglia which were immunostained with Iba-1 at 4 days post-ischemia, and tTG immunoreactivity in the microglia was also highest at 5 days post-ischemia. In Western blot analysis, tTG protein levels in the CA1 region after ischemia/reperfusion began to increase 3 days after ischemia/reperfusion and peaked 5 days after ischemia/reperfusion. The expression of tTG in PECAM-1-immunoreactive blood vessels may be associated with integrin regulation or transendothelial migration of leukocytes in the ischemic CA1 region. In this study, we also observed the effect of cystamine, a tTG inhibitor, against ischemic damage. Administration of cystamine protected in certain degree neuronal damage from ischemic damage in the CA1 region. These results suggest that tTG may be associated with neuronal death in the hippocampal CA1 region induced by ischemia/reperfusion.


Asunto(s)
Isquemia Encefálica/enzimología , Cistamina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Proteínas de Unión al GTP/metabolismo , Hipocampo/enzimología , Fármacos Neuroprotectores/uso terapéutico , Transglutaminasas/metabolismo , Análisis de Varianza , Animales , Benzoxazinas , Vasos Sanguíneos/enzimología , Western Blotting , Isquemia Encefálica/tratamiento farmacológico , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/antagonistas & inhibidores , Gerbillinae , Hipocampo/irrigación sanguínea , Hipocampo/efectos de los fármacos , Inmunohistoquímica , Masculino , Microglía/metabolismo , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oxazinas , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/fisiopatología , Transglutaminasas/antagonistas & inhibidores
19.
Apoptosis ; 13(9): 1184-94, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18670884

RESUMEN

Cisplatin, a chemotherapeutic drug that is widely used to treat various cancers, promotes ototoxicity at higher doses. In this study, the effect of epicatechin (EC) on cisplatin-induced hair cell death was investigated in a cochlear organ of Corti-derived cell line, HEI-OC1, and in vivo in zebrafish. Cisplatin promoted apoptosis and altered mitochondrial membrane potential (MMP) in HEI-OC1 cells. EC inhibited cisplatin-induced apoptosis and intracellular reactive oxygen species (ROS) generation. Labeling of zebrafish lateral line hair cells by the fluorescent dye YO-PRO1 was lost upon exposure to cisplatin, and EC protected against this cisplatin-induced loss of labeling in a dose-dependent manner. Scanning and transmission electron micrographs showed that treatment with EC protected against cisplatin-induced loss of kinocilium and stereocilia in zebrafish neuromasts. These results suggest that EC prevents cisplatin-induced ototoxicity by blocking ROS generation and by preventing changes in MMP.


Asunto(s)
Catequina/farmacología , Cisplatino/farmacología , Citoprotección/efectos de los fármacos , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/efectos de los fármacos , Animales , Caspasa 3/metabolismo , Catequina/química , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cisplatino/antagonistas & inhibidores , Células Ciliadas Auditivas/enzimología , Células Ciliadas Auditivas/ultraestructura , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra
20.
Cancer Lett ; 271(1): 140-52, 2008 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-18632202

RESUMEN

Hepatocyte growth factor (HGF) has recently attracted a considerable amount of attention as a stromal-derived mediator in tumor-stromal interactions, particularly because of its close involvement in cancer invasion and metastasis, and (-)-epigallocatechin-3-gallate (EGCG) can modulate the cell signaling associated with angiogenesis, metastasis, and migration of cancer cells. In the present study, we have investigated the effects of HGF on invasion and metastasis of hypopharyngeal carcinoma cells and the effect of EGCG on blocking HGF-induced invasion and metastasis in these cells. We found that HGF promoted the autophosphorylation of c-Met, HGF receptor, and that HGF-induced proliferation, colony dispersion, migration and invasion of tumors. We also observed that HGF enhanced the activity of matrix metalloproteinase (MMP)-9 and urokinase-type plasminogen activator (uPA) in hypopharyngeal carcinoma cells. In addition, HGF-induced the activation of Akt and Erk pathway as a downstreaming pathway of invasion. On the other hand, EGCG at physiologically relevant concentration (1 microM) suppressed HGF-induced tumor motility and MMP-9 and uPA activities, and the suppression of Akt and Erk pathway by EGCG was one of the downstream mechanisms to facilitate EGCG-induced anti-invasion effects. These results suggest that EGCG may serve as a therapeutic agent to inhibit HGF-induced invasion in hypopharyngeal carcinoma patients.


Asunto(s)
Catequina/análogos & derivados , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Neoplasias Hipofaríngeas/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Secuencia de Bases , Western Blotting , Catequina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cartilla de ADN , Inducción Enzimática , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Neoplasias Hipofaríngeas/enzimología , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA