Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
2.
Biol Pharm Bull ; 40(8): 1232-1239, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28769005

RESUMEN

Breast cancer is one of the most lethal malignancies in the world. Oxymatrine is the major effective and toxic alkaloid component which is derived from the root of Sophora flavescens AIT, a traditional Chinese medicine which is widely distributed in Asia and the Pacific Islands. In the current research study, we investigated the effects and mechanisms of action of oxymatrine on breast cancer cells. We demonstrated that the viability and single cell proliferation capability of MCF-7 and MDA-MB-231, two breast cancer cell lines which are widely used in in vitro study, were significantly suppressed in a time- and concentration-dependent manner. Furthermore, the cell cycle of breast cancer cells treated with oxymatrine was arrested at the S-phase of the cell cycle. Oxymatrine also triggered apoptosis in breast cancer cells by modulating apoptosis-related proteins, such as cleaved Caspase-3, cleaved Caspase-9 and poly(ADP-ribose)polymerase (PARP). The remarkable reduction in the ratio of Bcl-2/Bax was also observed in oxymatrine treated breast cancer cells. In conclusion, our research demonstrated that oxymatrine plays a critical role in suppressing carcinogenesis of breast cancer cells through cell cycle arrest and induction of mitochondria-mediated apoptosis, which suggests a promising application of this drug in breast cancer therapy.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Quinolizinas/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo
3.
Oncol Lett ; 13(3): 1223-1227, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28454238

RESUMEN

It has been reported previously that a dopamine receptor D2 (DRD2) antagonist was able to induce cancer cell apoptosis and that DRD2 was expressed at high levels in pituitary adenomas. However, the expression of DRD2 in gastric cancer and its correlation with the prognosis of patients with gastric cancer remain to be elucidated. In the present study, the expression of DRD2 in 84 paired gastric cancer tissues and respective adjacent non-cancerous tissues were detected using an immunohistochemical assay. The correlation between the expression of DRD2 and the with survival durations of the patients with gastric cancer was analyzed using Kaplan-Meier analysis. In addition, online resources were utilized to further analyze the correlation between the mRNA expression level of DRD2 and prognosis. The effect of the DRD2 antagonist, thioridazine, on the proliferation of the AGS gastric cancer cells was determined. The results of the present study showed that the percentage of gastric cancer cases with a high expression level of DRD2 (51.2%) was higher, compared with that of cases with a low expression level of DRD2 (39.3%). Patients with a higher expression of DRD2 had shorter survival durations. The online database analysis revealed that the expression of DRD2 was also inversely correlated with the prognosis of patients with gastric cancer. Furthermore, the DRD2 antagonist, thioridazine, inhibited the growth of AGS gastric cancer cells. In conclusion, as the expression of DRD2 was negatively correlated with survival durations in patients with gastric cancer, it may be considered as a prognosis marker in the future. Developing DRD2 antagonists may assist in increasing the efficiency of cancer therapy.

5.
Molecules ; 20(11): 20659-68, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26610442

RESUMEN

Gastric cancer is one of the most common malignant cancers, with high death rates, poor prognosis and limited treatment methods. Cyclovirobuxine D (CVB-D) is the main active component of the traditional Chinese medicine Buxus microphylla. In the present study, we test the effects of CVB-D on gastric cancer cells and the underlying mechanisms of action. CVB-D reduced cell viability and colony formation ability of MGC-803 and MKN28 cells in a time- and concentration-dependent manner. Flow cytometry showed that cell cycle of CVB-D treated cells was arrested at the S-phase. CVB-D also induced apoptosis in MGC-803 and MKN28 cells, especially early stage apoptosis. Furthermore, mitochondria membrane potential (Δψm) was reduced and apoptosis-related proteins, cleaved Caspase-3 and Bax/Bcl-2, were up-regulated in CVB-D-treated MGC-803 and MKN28 cells. Taken together, our studies found that CVB-D plays important roles in inhibition of gastric tumorigenesis via arresting cell cycle and inducing mitochondria-mediated apoptosis, suggesting the potential application of CVB-D in gastric cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neoplasias Gástricas/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neoplasias Gástricas/genética
6.
Drug Des Devel Ther ; 9: 3969-87, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26309394

RESUMEN

Gallbladder cancer (GBC), the most frequent malignancy of the biliary tract, is associated with high mortality and extremely poor prognosis. 20(S)-ginsenoside Rg3 (20(S)-Rg3) is a steroidal saponin with high pharmacological activity. However, the anticancer effect of 20(S)-Rg3 in human GBC has not yet been determined. In this study, we primarily found that 20(S)-Rg3 exposure suppressed the survival of both NOZ and GBC-SD cell lines in a concentration-dependent manner. Moreover, induction of cellular senescence and G0/G1 arrest by 20(S)-Rg3 were accompanied by a large accumulation of p53 and p21 as a result of murine double minute 2 (MDM2) inhibition. 20(S)-Rg3 also caused a remarkable increase in apoptosis via the activation of the mitochondrial-mediated intrinsic caspase pathway. Furthermore, intraperitoneal injection of 20(S)-Rg3 (20 or 40 mg/kg) for 3 weeks markedly inhibited the growth of xenografts in nude mice. Our results demonstrated that 20(S)-Rg3 potently inhibited growth and survival of GBC cells both in vitro and in vivo. 20(S)-Rg3 attenuated GBC growth probably via activation of the p53 pathway, and subsequent induction of cellular senescence and mitochondrial-dependent apoptosis. Therefore, 20(S)-Rg3 may be a potential chemotherapeutic agent for GBC therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Ginsenósidos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 10(1): e0114851, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25617627

RESUMEN

Baicalein, a widely used Chinese herbal medicine, has multiple pharmacological activities. However, the precise mechanisms of the anti-proliferation and anti-metastatic effects of baicalein on gallbladder cancer (GBC) remain poorly understood. Therefore, the aim of this study was to assess the anti-proliferation and anti-metastatic effects of baicalein and the related mechanism(s) on GBC. In the present study, we found that treatment with baicalein induced a significant inhibitory effect on proliferation and promoted apoptosis in GBC-SD and SGC996 cells, two widely used gallbladder cancer cell lines. Additionally, treatment with baicalein inhibited the metastasis of GBC cells. Moreover, we demonstrated for the first time that baicalein inhibited GBC cell growth and metastasis via down-regulation of the expression level of Zinc finger protein X-linked (ZFX). In conclusion, our studies suggest that baicalein may be a potential phytochemical flavonoid for therapeutics of GBC and ZFX may serve as a molecular marker or predictive target for GBC.


Asunto(s)
Carcinoma/patología , Regulación hacia Abajo , Medicamentos Herbarios Chinos/farmacología , Flavanonas/farmacología , Neoplasias de la Vesícula Biliar/patología , Factores de Transcripción de Tipo Kruppel/genética , Animales , Apoptosis/efectos de los fármacos , Carcinoma/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flavanonas/química , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Humanos , Masculino , Ratones Desnudos , Metástasis de la Neoplasia/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Cell Int ; 14(1): 96, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25383044

RESUMEN

BACKGROUND: Ursolic acid (UA), a plant extract used in traditional Chinese medicine, exhibits potential anticancer effects in various human cancer cell lines in vitro. In the present study, we evaluated the anti-tumoral properties of UA against gallbladder carcinoma and investigated the potential mechanisms responsible for its effects on proliferation, cell cycle arrest and apoptosis in vitro. METHODS: The anti-tumor activity of UA against GBC-SD and SGC-996 cells was assessed using MTT and colony formation assays. An annexin V/PI double-staining assay was used to detect cell apoptosis. Cell cycle changes were detected using flow cytometry. Rhodamine 123 staining was used to assess the mitochondrial membrane potential (ΔΨm) and validate UA's ability to induce apoptosis in both cell lines. The effectiveness of UA in gallbladder cancer was further verified in vivo by establishing a xenograft GBC model in nude mice. Finally, the expression levels of cell cycle- and apoptosis-related proteins were analyzed by western blotting. RESULTS: Our results suggest that UA can significantly inhibit the growth of gallbladder cancer cells. MTT and colony formation assays indicated dose-dependent decreases in cell proliferation. S-phase arrest was observed in both cell lines after treatment with UA. Annexin V/PI staining suggested that UA induced both early and late phases of apoptosis. UA also decreased ΔΨm and altered the expression of molecules regulating the cell cycle and apoptosis. In vivo study showed intraperitoneally injection of UA can significantly inhibited the growth of xenograft tumor in nude mice and the inhibition efficiency is dose related. Activation of caspase-3,-9 and PARP indicated that mitochondrial pathways may be involved in UA-induced apoptosis. CONCLUSIONS: Taken together, these results suggest that UA exhibits significant anti-tumor effects by suppressing cell proliferation, promoting apoptosis and inducing 7cell cycle arrest both in vitro and in vivo. It may be a potential agent for treating gallbladder cancer.

9.
World J Surg Oncol ; 12: 322, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25344327

RESUMEN

BACKGROUND: Gastric cancer is one of the most common malignancies and is a leading cause of cancer death worldwide. Surgery is the most effective and successful method of treatment for gastric cancer, and systematic lymph node (LN) dissection is unquestionably the most effective procedure for treating LN metastases of gastric cancer. Systematic lymphadenectomy is the most important part of curative resection, but lymphadenectomy is also the most difficult procedure in gastric cancer surgery. The aim of this study is to report our three-step method for lymphadenectomy in gastric cancer. METHODS: In this study, the lymph node stations and groups were defined according to the 13th edition of the Japanese Classification for Gastric Carcinoma. The authors' novel, simplified method consists of three steps: (1) the Kocher maneuver and dissection of the greater omentum together with the anterior sheet of the mesocolon, (2) dissection of the lesser omentum, and (3) lymphadenectomy following the main vessels. We primarily used Peng's multifunctional operative dissector, which combines four different functions (cutting, separating, aspirating and coagulating). Our systematic lymphadenectomy included three steps, and the main procedure started from right to left and in the caudal to cranial direction. RESULTS: A total of 830 consecutive patients underwent our three-step-method systematic lymphadenectomy in advanced gastric cancer surgery. The mean operation time was 146 minutes, and the mean blood loss was 248 ml. The median postoperative hospital stay was 10.9±4.8 days. The median number of examined LN was 31.6 (range 17 to 72) per patient, and the median number of metastatic LN was 5.6 (range 0 to 42) per patient. The overall incidence of postoperative complications was 10.6%, and the rate of hospital death was 0.9%. The overall three-year survival rate was 52.6%. CONCLUSIONS: Our three-step method for lymphadenectomy is easy to perform and is a useful procedure for gastric cancer surgery.


Asunto(s)
Disección/métodos , Escisión del Ganglio Linfático/métodos , Neoplasias Gástricas/cirugía , Legrado , Disección/instrumentación , Gastrectomía , Humanos , Escisión del Ganglio Linfático/instrumentación , Metástasis Linfática , Mesocolon/cirugía , Epiplón/cirugía , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Succión , Tasa de Supervivencia , Resultado del Tratamiento
10.
Cancer Lett ; 355(2): 201-9, 2014 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-25218593

RESUMEN

The transcriptional coactivator Yes-associated protein 1 (YAP1), a key regulator of cell proliferation and organ size in vertebrates, has been implicated in various malignancies. However, little is known about the expression and biological function of YAP1 in human gallbladder cancer (GBC). In this study we examined the clinical significance and biological functions of YAP1 in GBC and found that nuclear YAP1 and its target gene AXL were overexpressed in GBC tissues. We also observed a significant correlation between high YAP1 and AXL expression levels and worse prognosis. The depletion of YAP1 using lentivirus shRNAs significantly inhibited cell proliferation by inducing cell cycle arrest in S phase in concordance with the decrease of CDK2, CDC25A, and cyclin A, and resulted in increased cell apoptosis and invasive repression in GBC cell lines in vitro. Furthermore, knockdown of YAP1 also inhibited tumor growth in vivo. Additionally, we demonstrated that the activation of the AXL/MAPK pathway was involved in the oncogenic functions of YAP1 in GBC. These results demonstrated that YAP1 is a putative oncogene and represents a prognostic marker and potentially a novel therapeutic target for GBC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Vesícula Biliar/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/fisiología , Activación Enzimática , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/genética , Oncogenes , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Fase S/fisiología , Factores de Transcripción , Proteínas Señalizadoras YAP , Tirosina Quinasa del Receptor Axl
11.
Molecules ; 19(8): 11350-65, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25090123

RESUMEN

Gallbladder cancer is the most common malignant tumor of the biliary tract, and this condition has a rather dismal prognosis, with an extremely low five-year survival rate. To improve the outcome of unresectable and recurrent gallbladder cancer, it is necessary to develop new effective treatments and drugs. The purpose of the present study was to evaluate the effects of cordycepin on human gallbladder cells and uncover the molecular mechanisms responsible for these effects. The Cell Counting Kit-8 (CCK-8) and colony formation assays revealed that cordycepin affected the viability and proliferation of human gallbladder cancer cells in a dose- and time-dependent manner. Flow cytometric analysis showed that cordycepin induced S phase arrest in human gallbladder cancer cell lines(NOZ and GBC-SD cells). Cordycepin-induced apoptosis was observed using an Annexin V/propidium iodide (PI) double-staining assay, and the mitochondrial membrane potential (ΔΨm) decreased in a dose-dependent manner. Additionally, western blot analysis revealed the upregulation of cleaved-caspase-3, cleaved-caspase-9, cleaved-PARP and Bax and the downregulation of Bcl-2, cyclin A and Cdk-2 in cordycepin-treated cells. Moreover, cordycepin inhibited tumor growth in nude mice bearing NOZ tumors. Our results indicate that this drug may represent an effective treatment for gallbladder carcinoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Desoxiadenosinas/farmacología , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Animales , Antineoplásicos/química , Caspasa 3/genética , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxiadenosinas/química , Modelos Animales de Enfermedad , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Estructura Molecular , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Tumour Biol ; 35(11): 10931-41, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25085580

RESUMEN

Bufalin, a major digoxin-like immunoreactive component of the Chinese medicine Chan Su, has been shown to exert a potential for anticancer activity against various human cancer cell lines in vitro. However, no detailed studies have so far been reported on its action on human gallbladder carcinoma cells. In this study, bufalin remarkably inhibited growth in human gallbladder cancer cells by decreasing cell proliferation, inducing cell cycle arrest and apoptosis in a dose-dependent manner. Bufalin also disrupted the mitochondrial membrane potential (ΔΨm) and regulated the expression of cell cycle and apoptosis regulatory molecules. Activation of caspase-9 and the subsequent activation of caspase-3 indicated that bufalin may be inducing mitochondria apoptosis pathways. Intraperitoneal injection of bufalin for 3 weeks significantly inhibited the growth of gallbladder carcinoma (GBC-SD) xenografts in athymic nude mice. Taken together, the results indicate that bufalin may be a potential agent for the treatment of gallbladder cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bufanólidos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/patología , Animales , Western Blotting , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/metabolismo , Humanos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Molecules ; 19(9): 13235-50, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25165862

RESUMEN

Gallbladder cancer, with high aggressivity and extremely poor prognosis, is the most common malignancy of the bile duct. The main objective of the paper was to investigate the effects of schisandrin B (Sch B) on gallbladder cancer cells and identify the mechanisms underlying its potential anticancer effects. We showed that Sch B inhibited the viability and proliferation of human gallbladder cancer cells in a dose-, time -dependent manner through MTT and colony formation assays, and decrease mitochondrial membrane potential (ΔΨm) at a dose-dependent manner through flow cytometry. Flow cytometry assays also revealed G0/G1 phase arrest and apoptosis in GBC-SD and NOZ cells. Western blot analysis of Sch B-treated cells revealed the upregulation of Bax, cleaved caspase-9, cleaved caspase-3, cleaved PARP and downregulation of Bcl-2, NF-κB, cyclin D1 and CDK-4. Moreover, this drug also inhibited the tumor growth in nude mice carrying subcutaneous NOZ tumor xenografts. These data demonstrated that Sch B induced apoptosis in gallbladder cancer cells by regulating apoptosis-related protein expression, and suggests that Sch B may be a promising drug for the treatment of gallbladder cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Lignanos/administración & dosificación , Compuestos Policíclicos/administración & dosificación , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclooctanos/administración & dosificación , Neoplasias de la Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , FN-kappa B/biosíntesis , Proteínas de Neoplasias/biosíntesis
14.
Int J Clin Pharmacol Ther ; 52(10): 900-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25074868

RESUMEN

OBJECTIVE: The aims of this study were to observe the apoptosis effects of tetrandrine on human gallbladder carcinoma cell line (SGC-996), and to explore its related mechanism. METHODS: First, the anti-proliferative activities of tetrandrine on SGC-996 cells were determined by using the MTT assays. Then, cell cycle changes were detected by flow cytometry analysis. The apoptosis of cells was detected by the annexin V/propidium iodide double-staining assay. Detection of mitochondrial membrane potential was used to validate the ability of tetrandrine on inducing apoptosis. Finally, the expressions of the apoptosis-related proteins (caspase-3, PARP, Bcl-2, and Bax) were analyzed by western blot. Statistical analyses were performed using the Student’s t-test for comparison of the results obtained from cells with or without treatment of tetrandrine. RESULTS: The MTT assay revealed a significant inhibition of cell proliferation in a dose- and time-dependent manner. Cells treated with tetrandrine were arrested at the S phase, according to the flow cytometric analysis. Tetrandrine produced a dose-dependent increase in the apoptotic cell population compared with control cells. Tetrandrine can also affect mitochondrial function by changing the mitochondrial membrane potential. Furthermore, western blot assay demonstrated that the tetrandrine induced apoptosis in SGC-996 cells by regulating the ratio of Bcl-2/Bax and activating the expression of cleaved caspase-3. CONCLUSIONS: The results indicate that tetrandrine may be a potential agent for the treatment of gallbladder carcinoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Bencilisoquinolinas/farmacología , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Caspasa 3/análisis , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias de la Vesícula Biliar/patología , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteína X Asociada a bcl-2/análisis
15.
Nat Genet ; 46(8): 872-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24997986

RESUMEN

Individuals with gallbladder carcinoma (GBC), the most aggressive malignancy of the biliary tract, have a poor prognosis. Here we report the identification of somatic mutations for GBC in 57 tumor-normal pairs through a combination of exome sequencing and ultra-deep sequencing of cancer-related genes. The mutation pattern is defined by a dominant prevalence of C>T mutations at TCN sites. Genes with a significant frequency (false discovery rate (FDR)<0.05) of non-silent mutations include TP53 (47.1%), KRAS (7.8%) and ERBB3 (11.8%). Moreover, ErbB signaling (including EGFR, ERBB2, ERBB3, ERBB4 and their downstream genes) is the most extensively mutated pathway, affecting 36.8% (21/57) of the GBC samples. Multivariate analyses further show that cases with ErbB pathway mutations have a worse outcome (P=0.001). These findings provide insight into the somatic mutational landscape in GBC and highlight the key role of the ErbB signaling pathway in GBC pathogenesis.


Asunto(s)
Carcinoma/genética , Receptores ErbB/genética , Exoma , Neoplasias de la Vesícula Biliar/genética , Mutación , Recurrencia Local de Neoplasia/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma/enzimología , Línea Celular , Línea Celular Tumoral , Femenino , Neoplasias de la Vesícula Biliar/enzimología , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/enzimología , Transducción de Señal/genética
16.
BMC Cancer ; 14: 217, 2014 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-24655726

RESUMEN

BACKGROUND: Gallbladder cancer is the most frequent malignancy of the bile duct with high aggressive and extremely poor prognosis. The main objective of the paper was to investigate the inhibitory effects of oridonin, a diterpenoid isolated from Rabdosia rubescens, on gallbladder cancer both in vitro and in vivo and to explore the mechanisms underlying oridonin-induced apoptosis and cell cycle arrest. METHODS: The anti-tumor activity of oridonin on SGC996 and NOZ cells was assessed by the MTT and colony forming assays. Cell cycle changes were detected by flow cytometric analysis. Apoptosis was detected by annexin V/PI double-staining and Hoechst 33342 staining assays. Loss of mitochondrial membrane potential was observed by Rhodamine 123 staining. The in vivo efficacy of oridonin was evaluated using a NOZ xenograft model in athymic nude mice. The expression of cell cycle- and apoptosis-related proteins in vitro and in vivo was analyzed by western blot analysis. Activation of caspases (caspase-3, -8 and -9) was measured by caspases activity assay. RESULTS: Oridonin induced potent growth inhibition, S-phase arrest, apoptosis, and colony-forming inhibition in SGC996 and NOZ cells in a dose-dependent manner. Intraperitoneal injection of oridonin (5, 10, or 15 mg/kg) for 3 weeks significantly inhibited the growth of NOZ xenografts in athymic nude mice. We demonstrated that oridonin regulated cell cycle-related proteins in response to S-phase arrest by western blot analysis. In contrast, we observed inhibition of NF-κB nuclear translocation and an increase Bax/Bcl-2 ratio accompanied by activated caspase-3, caspase-9 and PARP-1 cleavage after treatment with oridonin, which indicate that the mitochondrial pathway is involved in oridonin-mediated apoptosis. CONCLUSIONS: Oridonin possesses potent anti-gallbladder cancer activities that correlate with regulation of the mitochondrial pathway, which is critical for apoptosis and S-phase arrest. Therefore, oridonin has potential as a novel anti-tumor therapy for the treatment of gallbladder cancer.


Asunto(s)
Antineoplásicos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Neoplasias de la Vesícula Biliar/patología , Mitocondrias/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias de la Vesícula Biliar/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Experimentales , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Biol Ther ; 15(6): 806-14, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24658096

RESUMEN

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNA (lncRNA), is associated with metastasis and is an independent prognostic factor for lung cancer. Recent studies have demonstrated that MALAT1 plays an important role in other malignancies. However, little is known about the role of MALAT1 in gallbladder carcinoma (GBC), which is the most common cancer of the biliary tract and has an extremely poor prognosis. In this study, we focused on the expression, biological functions and mechanism of MALAT1 in GBC and found that MALAT1 was significantly upregulated in GBC tissues compared with corresponding non-cancerous tissues. Knockdown of MALAT1 in GBC cell lines using lentivirus-mediated RNA interference significantly inhibited the proliferation and metastasis of the GBC cells both in vitro and in vivo. Furthermore, ERK/MAPK pathway was found to be inactivated in the GBC cell lines after MALAT1 knockdown. These results indicated that MALAT1 might serve as an oncogenic lncRNA that promotes proliferation and metastasis of GBC and activates the ERK/MAPK pathway.


Asunto(s)
Adenocarcinoma/secundario , Movimiento Celular , Proliferación Celular , Neoplasias de la Vesícula Biliar/patología , Sistema de Señalización de MAP Quinasas , ARN Largo no Codificante/fisiología , Adenocarcinoma/metabolismo , Línea Celular Tumoral , Femenino , Neoplasias de la Vesícula Biliar/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Persona de Mediana Edad
18.
Oncol Rep ; 31(5): 2107-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24604290

RESUMEN

Thioridazine, an antipsychotic drug, has been reported to induce apoptosis in various types of cancer cells, with specificity on targeting cancer stem cells (CSCs). However, whether it elicits anticancer effects in gastric cancer has never been reported. In the present study, we examined the ability of thioridazine to induce cell death in the gastric cancer cell lines NCI-N87 and AGS, and detected its in vivo tumor inhibition capacity. Thioridazine elicited cytotoxic effects on NCI-N87 and AGS cells in a dose-dependent manner, and inhibited the colony formation abilitiy of the NCI-N87 and AGS cells. Thioridazine treatment induced nuclear fragmentation, increased the proportion of sub-G1 phase cells, and elevated the percentage of Annexin V-positive cells, suggesting the occurrence of apoptosis. Moreover, thioridazine induced gastric cancer cell apoptosis in a caspase-dependent manner, as shown by a decrease in the precursors of casapse-9, caspase-8 and caspase-3, and by the ability of the caspase inhibitor Z-VAD-FMK to reverse the cytotoxic effect of thioridazine. JC-1 staining further revealed that thioridazine induced gastric cancer cell apoptosis via the mitochondrial pathway. In addition, thioridazine pretreatment inhibited the growth of NCI-N87 cell-derived tumors. The present study demonstrated that the antipsychotic drug thioridazine possesses anti-gastric cancer ability through in vitro and in vivo experiments, suggesting thioridazine as a potential drug in gastric cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Antipsicóticos/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Tioridazina/uso terapéutico , Clorometilcetonas de Aminoácidos/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 8/biosíntesis , Caspasa 9/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo
19.
Anticancer Agents Med Chem ; 14(8): 1136-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24568162

RESUMEN

Baicalin, the main active ingredient in the Scutellaria baicalensis (SB), is prescribed for the treatment of various inflammatory diseases and tumors in clinics in China. In the present study, we evaluated the antitumor activity of baicalin for gallbladder carcinoma and the underlying mechanisms both in vitro and in vivo. Our results indicate that baicalin induced potent growth inhibition, cell cycle arrest, apoptosis and colony-formation inhibition in a dose-dependent manner in vitro. We observed inhibition of NF-κB nuclear translocation, up-regulation of Bax and down-regulation of Bcl-2, as well as increased caspase-3 and caspase-9 expression after baicalin treatment in vitro and in vivo, which indicates that the mitochondrial pathway was involved in baicalin-induced apoptosis. In addition, daily intraperitoneally injection of baicalin (15, 30 and 60 mg/kg) for 21 days significantly inhibited the growth of NOZ cells xenografts in nude mice, which improved the survival of baicalin-treated mice. In summary, baicalin exhibited a significant anti-tumor effect by suppressing cell proliferation, promoting apoptosis, and inducing cell cycle arrest in vitro, and by suppressing tumor growth and improving survival in vivo, which suggested that baicalin represents a novel therapeutic option for gallbladder carcinoma.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma/metabolismo , Flavonoides/farmacología , Neoplasias de la Vesícula Biliar/metabolismo , Mitocondrias/metabolismo , Animales , Antineoplásicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Ciclina B1/metabolismo , Ciclina D1/metabolismo , Flavonoides/uso terapéutico , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Neoplasias de la Vesícula Biliar/patología , Xenoinjertos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Desnudos , Necrosis , Transducción de Señal
20.
Molecules ; 19(2): 2612-28, 2014 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-24566325

RESUMEN

Gallbladder carcinoma is the most common malignancy of the biliary tract, with a very low 5-year survival rate and extremely poor prognosis. Thus, new effective treatments and drugs are urgently needed for the treatment of this malignancy. In this study, for the first time we investigated the effects of triptolide on gallbladder cancer cells and identified the mechanisms underlying its potential anticancer effects. The MTT assay showed that triptolide decreased cell viability in a dose- and time-dependent manner. The results of the colony formation assay indicated that triptolide strongly suppressed colony formation ability in GBC-SD and SGC-996 cells. Flow cytometric analysis revealed that triptolide induced S phase arrest in gallbladder cancer cells. In addition, triptolide induced apoptosis, as shown by the results of annexin V/propidium iodide double-staining and Hoechst 33342 staining. Furthermore, triptolide decreased mitochondrial membrane potential (ΔΨm) in a dose-dependent manner. Finally, western blot analysis of triptolide-treated cells revealed the activation of caspase-3, caspase-9, PARP, and Bcl-2; this result demonstrated that triptolide induced apoptosis in gallbladder cancer cells by regulating apoptosis-related protein expression, and suggests that triptolide may be a promising drug to treat gallbladder carcinoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Diterpenos/química , Neoplasias de la Vesícula Biliar/tratamiento farmacológico , Fenantrenos/química , Fase S/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Diterpenos/administración & dosificación , Compuestos Epoxi/administración & dosificación , Compuestos Epoxi/química , Neoplasias de la Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Fenantrenos/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...