Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Environ Manage ; 367: 122022, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39106802

RESUMEN

Identifying the driving forces of surface water quality variations is crucial for urban environmental management, especially in densely populated regions. Statistic mapping is an approach that allows researchers to directly explore the response of surface water quality to potential drivers. Conventionally, these methods encounter a mixture of issues, including nonlinear relationships and information on multiple time-scale, caused by disparities in the influencing frequencies and degrees of driving factors. In this research, a nonlinear direct-mapping approach was developed to quantitatively analyze the driving force of surface water quality under multiple time scales. This approach separated the fluctuation and trend information from water quality data and then established a direct-mapping relationship, thereby achieving the visible multilayer structure containing both linear and nonlinear information from the time scale. Typical water pollutants including total nitrogen (TN) and total phosphorus (TP) in the Pearl River Delta (PRD), were used to verify the methodology and compare its ability to analyze driving forces with traditional statistic approaches. The results demonstrated that this approach could establish a visual multilayer mapping structure with strong interpretability, which effectively captured the contained nonlinear information, thus improving the fitting degree by 12.43% compared with traditional methods. Moreover, it successfully identified the dominant driving forces of TN and TP in the PRD as human activities related to NO2 and PM and natural factors. Its application in the changing environment demonstrated a potentially increased risk of TP in the PRD under multiple scenarios. Overall, this approach could serve as a reliable reference for pollution early warning in the short term and for industrial structure adjustment planning in the long term.


Asunto(s)
Monitoreo del Ambiente , Nitrógeno , Fósforo , Calidad del Agua , Nitrógeno/análisis , Humanos , Monitoreo del Ambiente/métodos , Fósforo/análisis , Ríos/química
2.
Cell Mol Biol (Noisy-le-grand) ; 67(2): 8-13, 2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34817344

RESUMEN

The current study was designed to unveil the anticancer effects of naringenin against breast cancer MDA-MB-231 cells. Cytotoxic effects were estimated via MTT viability assay. Clonogenic assay was performed to assess clonogenic potential of MDA-MB-231 cells. Apoptosis was examined via AO/EB staining, quantified via annexin V/PI staining and western blotting was performed to monitor apoptosis allied protein expressions. Cell cycle was analyzed through flow cytometric analysis. Transwell chambers assay was executed for determination of cell migration and cell invasion tendency of MDA-MB-231 breast cancer cells. Results indicated significant anticancer potential of naringenin drug against MDA-MB-231 cells. On evaluation of cell proliferation rate of breast cancer cells by MTT assay, it was observed that naringenin inhibited proliferation rate in dose as well as time dependent manner. AO/EB staining assay revealed potential morphological changes indicating apoptotic cell death. Annexin V/PI staining assay revealed increased apoptotic cell percentage with increased drug doses. The apoptosis inducing potential of naringenin drug was observed to be mediated via caspase activation. Flow cytometric analysis predicted cell cycle arrest at G2/M phase of cell cycle. Further cell migration as well as cell invasion tendency of MDA-MB-231 cells was reduced to minimum upon application of naringenin drug.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Caspasas/metabolismo , Flavanonas/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Antineoplásicos Fitogénicos/química , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Flavanonas/química , Humanos , Estructura Molecular , Metástasis de la Neoplasia , Factores de Tiempo
3.
J Cell Mol Med ; 19(3): 630-41, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25598321

RESUMEN

ß-elemene, a compound derived from Rhizoma zedoariae, is a promising new plant-derived drug with broad-spectrum anticancer activity. However, the underlying mechanism by which this agent inhibits human lung cancer cell growth has not been well elucidated. In this study, we showed that ß-elemene inhibits human non-small cell lung carcinoma (NSCLC) cell growth, and increased phosphorylation of ERK1/2, Akt and AMPKα. Moreover, ß-elemene inhibited expression of DNA methyltransferase 1 (DNMT1), which was not observed in the presence of the specific inhibitors of ERK (PD98059) or AMPK (compound C). Overexpression of DNMT1 reversed the effect of ß-elemene on cell growth. Interestingly, metformin not only reversed the effect of ß-elemene on phosphorylation of Akt but also strengthened the ß-elemene-reduced DNMT1. In addition, ß-elemene suppressed Sp1 protein expression, which was eliminated by either ERK1/2 or AMPK inhibitor. Conversely, overexpression of Sp1 antagonized the effect of ß-elemene on DNMT1 protein expression and cell growth. Taken together, our results show that ß-elemene inhibits NSCLC cell growth via ERK1/2- and AMPKα-mediated inhibition of transcription factor Sp1, followed by reduction in DNMT1 protein expression. Metformin augments the effect of ß-elemene by blockade of Akt signalling and additively inhibition of DNMT1 protein expression. The reciprocal ERK1/2 and AMPKα signalling pathways contribute to the overall responses of ß-elemene. This study reveals a potential novel mechanism by which ß-elemene inhibits growth of NSCLC cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Sesquiterpenos/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metformina/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción Sp1/metabolismo
4.
Mol Cell Biochem ; 402(1-2): 63-74, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25547067

RESUMEN

Hepatocellular carcinoma (HCC), the major histological subtype of primary liver cancer, remains one of the most common malignancies worldwide. Due to the complicated pathogenesis of this malignancy, the outcome for comprehensive treatment is limited. Chinese herbal medicine (CHM) is emerging as a promising choice for its multi-targets and coordinated intervention effects against HCC. Ursolic acid (UA), a natural pentacyclic triterpenoid carboxylic acid found in CHM, exerts anti-tumor effects and is emerging as an effective compound for cancer prevention and therapy. However, the molecular mechanisms underlying the action of UA remain largely unknown. In this study, we showed that UA inhibited the growth of HCC cells and induced apoptosis in the dose- and time-dependent fashion. Furthermore, we found that UA induced phosphorylation of AMP-activated protein kinase alpha (AMPKα) and suppressed the protein expression of DNA methyltransferase 1 (DNMT1) in the dose-dependent manner. The inhibitor of AMPK, compound C blocked, while an activator of AMPK, metformin augmented the effect of UA on DNMT1 expression. In addition, UA suppressed the expression of transcription factor Sp1. Conversely, overexpression of Sp1 reversed the effect of UA on DNMT1 expression and cell growth. Collectively, our results show for the first time that UA inhibits growth of HCC through AMPKα-mediated inhibition of Sp1; this in turn results in inhibition of DNMT1. This study reveals a potential novel mechanism by which UA controls growth of HCC cells and suggests that DNMT1 could be novel target for HCC chemoprevention and treatment.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Antineoplásicos Fitogénicos/farmacología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Triterpenos/farmacología , Apoptosis , Carcinoma Hepatocelular , Proliferación Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Concentración 50 Inhibidora , Metformina/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Factor de Transcripción Sp1/metabolismo , Ácido Ursólico
5.
Mol Cancer ; 13: 149, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24925061

RESUMEN

BACKGROUND: Peroxisome proliferator-activated receptors gamma (PPARγ) ligands have been shown to inhibit the growth of non-small cell lung cancer (NSCLC) cells. However, the mechanisms underlying this effect remain incompletely elucidated. METHODS: Cell proliferation and apoptosis were measured by cell viability, MTT and caspase3/7 activity assays. Phosphorylation/protein expression and gene silence/overexpression of AMPKα, phosphoinositide-dependent protein kinase 1 (PDK1), Egr-1 and PPARγ were performed by Western blot and siRNA/transfection assays. Dual-Luciferase Reporter Kit was used to measure the PPAR response elements (PPRE) reporter and PDK1 promoter activities, and ChIP assay was used to detect the Egr-1 protein binding to the DNA site in the PDK1 gene promoter. RESULTS: We found that ciglitazone, one synthetic PPARγ ligand, inhibited growth and induced apoptosis of NSCLC cells through decreased expression of PDK1, which was not blocked by GW9662 (a specific PPARγ antagonist). Overexpression of PDK1 overcame the effect of ciglitazone on cell growth and caspase 3/7 activity. Ciglitazone increased the phosphorylation of AMPKα and c-Jun N-terminal kinase (JNK), and the inhibitor of AMPK (compound C), but not JNK (SP600125), reversed the effect of ciglitazone on PDK1 protein expression. Ciglitazone reduced PDK1 gene promoter activity, which was not observed in cells exposed to compound C, but not silenced of PPARγ siRNA. Combination of ciglitazone and metformin further reduced PDK1 expression and promoter activity. Furthermore, we showed that ciglitazone induced the protein expression of Egr-1, which was not observed in cells silencing of AMPKα. Moreover, silencing of Egr-1 abrogated the effect of ciglitazone on PDK1 promoter activity and cell growth. On the contrary, overexpression of Egr-1 enhanced the effect of ciglitazone on PDK1 gene promoter activity. ChIP assays demonstrated that ciglitazone induced Egr-1 protein bind to the specific DNA site in the PDK1 gene promoter. CONCLUSION: Collectively, our results demonstrate that ciglitazone inhibits PDK1 expression through AMPKα-mediated induction of Egr-1 and Egr-1 binding to the specific DNA site in the PDK1 gene promoter, which is independent of PPARγ. Activation of AMPKα by metformin enhances the effect of ciglitazone. In turn, this leads to inhibition of NSCLC cell proliferation.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Neoplasias Pulmonares/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Metformina/administración & dosificación , PPAR gamma/genética , Regiones Promotoras Genéticas , Transducción de Señal , Tiazolidinedionas/administración & dosificación
6.
Tumour Biol ; 35(8): 8169-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24845028

RESUMEN

Solamargine (SM), a major steroidal alkaloid glycoside extracted from a traditional Chinese medicinal herb, Solanum nigrum L. (SNL), has been shown to inhibit growth and induce apoptosis of various cancer cells. However, the molecular mechanisms underlying this are poorly understood. In this study, we showed that SM inhibited growth and induced apoptosis of non-small-cell lung cancer (NSCLC) cells in a time- and dose-dependent manner. To further explore this, we found that SM increased phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK) in a time-dependent fashion. SM also inhibited phosphorylation and protein expression of signal transducer and activator of transcription 3 (Stat3), a transcription factor, which was abrogated by the SB203580, a specific inhibitor of p38 MAPK. In addition, SM induced protein expression of p21, one of cyclin-dependent kinase inhibitors, and this was not observed in cell overexpression of Stat3 or cells treated with SB203580. Finally, while silencing of Stat3 had no further effect, exogenous expression of Stat3 overcame the effect of SM on cell proliferation. Collectively, our results show that SM inhibits proliferation and induces apoptosis in lung cancer cells through p38 MAPK-mediated suppression of phosphorylation and protein expression of Stat3, followed by inducing Stat3 downstream effector p21. This unveils a potential new mechanism by which SM inhibits growth of human lung cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Alcaloides Solanáceos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Humanos , Neoplasias Pulmonares/patología , Fosforilación , Factor de Transcripción STAT3/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Environ Sci Pollut Res Int ; 30(2): 3252-3269, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35943654

RESUMEN

Accurate carbon price forecasting is of great significance to the operation of carbon financial markets. However, limited by the non-linearity and non-stationarity of the carbon price, the accurate and reliable predictions are difficult. To address the issue of applicability and accuracy, a novel carbon price hybrid model based on decomposition, entropy, and machine learning methods is proposed, named as CEEMDAN-PE-LSTM-RVM. Adopting the advanced structure (i.e., the prediction under classification), the proposed model owns reliable performance in face of the cases with different complexity. Furthermore, the relationship between the data feature and prediction accuracy is discussed to provide a benchmark for judging the reliability of the prediction, in which the chaos degree is introduced as a feature to characterize carbon price quantitatively. The performance of the proposed model is evaluated through historical data of four representative carbon prices. The results show that the average MAPE and RMSE of the proposed model achieve 1.7027 and 0.7993, respectively, which is significantly greater than others; the proposed model owns great robustness, which is less affected by the complexity of predicted objects. Thus, the proposed model provides a reliable tool for carbon financial markets.


Asunto(s)
Benchmarking , Carbono , Reproducibilidad de los Resultados , Entropía , Aprendizaje Automático , Predicción
8.
Zhen Ci Yan Jiu ; 42(2): 163-7, 2017 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-29071967

RESUMEN

OBJECTIVE: To observe the clinical efficacy of modified acupuncture at Renying point (ST 9) for patients with cervical spondylosis of vertebral artery type and its influence on velocity of cervical blood flow. METHODS: Fifty-nine cases of vertebral artery type cervical spondylosis were randomly divided into control group (n=30) and treatment group (n=29). Both groups were acupunctured at ST 9, with routine acupuncture technique used in the control group and modified technique in the treatment group, respectively. All cases received two courses of treatment, each course covered consecutive 6 once-per-day treatments. Before and after treatment, transcranial Doppler (TCD) was used to measure the systolic peak blood flow velocity (Vs) of left vertebral artery (LVA), right vertebral artery (RVA) and basilar artery (BA), and the scores of "cervical vertigo symptoms and functional assessment scale" (CVSFAS) were also assessed, separately. RESULTS: CVSFAS scoring, Vs of LVA, RVA and BA after treatment showed significant improvement compared with those before treatment (P<0.01, P<0.05). The efficacy of the treatment group in the above mentioned indexes was superior to that of the control group (P<0.05). The total effective rate of the treatment group was 93.1% (27/29), superior to 70.0% (21/30) of the control group (P<0.05). CONCLUSIONS: The modified acupuncture method at ST 9 is clinically effective in the treatment of cervical spondylosis of vertebral artery type via increasing the Vs of vertebral-basilar artery, improving the local blood circulation and relieving pain.


Asunto(s)
Terapia por Acupuntura , Espondilosis/terapia , Arteria Vertebral/fisiopatología , Puntos de Acupuntura , Adulto , Velocidad del Flujo Sanguíneo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Espondilosis/fisiopatología , Adulto Joven
9.
J Ethnopharmacol ; 181: 172-81, 2016 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-26850724

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Xiaoji decoction (XJD), which was considered as a Chinese herbal prescription, has been used for cancer treatment, especially lung cancer, for decades to improve quality of life and prolong the patient survival. However, the molecular mechanisms underlying the therapeutic potential have not been well elucidated. MATERIALS AND METHODS: The cell viability was examined by MTT assays. The phosphorylation and expression of AMP-activated protein kinase alpha (AMPKα), DNA methyltransferase 1 (DNMT1) and transcription factor Sp1 proteins were assessed by Western Blot. Exogenous expression of Sp1 and DNMT1 were performed by transient transfection methods. The effects of XJD on the growth of xenograft tumors were evaluated by in vivo bioluminescence imaging. RESULTS: We showed that XJD inhibited growth of human non small cell lung cancer (NSCLC) cells in vitro. We also found that XJD increased phosphorylation of AMPKα and inhibited protein expression of DNTM1, the latter was not observed in the presence of the inhibitor of AMPK (compound C). Overexpression of DNTM1 reversed the effect of XJD on cell growth. In addition, XJD decreased Sp1 protein expression, which was eliminated by compound C. Conversely, exogenous expressed Sp1 abrogated XJD-inhibited DNTM1 protein expression. Interestingly, exogenous expression of DNMT1 feedback antagonized the XJD-induced phosphorylation of AMPKα. In in vivo studies, we found that XJD inhibited tumor growth in xenograft nude mice model, which was accompanied by induction of phosphorylation of AMPKα and suppression of DNMT1 protein from xenograft tumors. CONCLUSION: Our results show that XJD inhibits NSCLC cell growth via AMPKα-mediated inhibition of transcription of Sp1, followed by the reduction of DNMT1 expression both in vitro and in vivo. The negative feedback regulation loop of AMPKα further demonstrates the critical role of DNMT1 in mediating the overall effects of XJD in this process. This study unveils novel molecular mechanism by which XJD controls NSCLC cell growth.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , ADN/metabolismo , Medicamentos Herbarios Chinos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Metiltransferasas/metabolismo , Factor de Transcripción Sp1/metabolismo , Células A549 , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Cell Signal ; 27(7): 1469-77, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25889897

RESUMEN

Despite the anti-cancer effect of emodin observed in several cancers, the underlying molecular mechanism remains to be elucidated. In this study, we showed that emodin-inhibited NSCLC cell growth and increased phosphorylation of AMPKα and ERK1/2. In addition, emodin-inhibited ILK protein expression. The overexpression of ILK reversed the effect of emodin on cell growth inhibition. Furthermore, the blockade of AMPK by compound C abrogated, while metformin, an activator of AMPK, strengthened the effect of emodin on the inhibition of ILK expression. Interestingly, the inhibitor of MAPK extracellular signaling-regulated kinase (ERK) kinase (MEK)/ERK1/2 (PD98059) attenuated emodin-induced phosphorylation of AMPKα. Moreover, emodin reduced the protein expression of Sp1 and AP-1 subunit c-Jun. Exogenous expression of Sp1 and c-Jun diminished emodin-reduced ILK protein expression. Emodin suppressed ILK promoter activity, which was not observed in cells overexpression of Sp1 and treated with compound C. Intriguingly, exogenous expression of c-Jun overcame the emodin-inhibited Sp1 protein expression. Collectively, our results demonstrate that emodin inhibits ILK expression through AMPKα-mediated reduction of Sp1 and c-Jun. Metformin enhances the effects of emodin. Exogenous expression of Sp1 and c-Jun resists emodin-inhibited ILK promoter activity and protein expression. In addition, the overexpression of c-Jun diminishes emodin-induced AMPKα signaling. Thus, the crosstalk of AMPKα and MEK/ERK1/2 signaling and the reciprocal interaction between Sp1 and c-Jun proteins contribute to the overall responses of emodin. This novel signaling axis may be a therapeutic potential for prevention and treatment of NSCLC.


Asunto(s)
Emodina/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Hipoglucemiantes/toxicidad , Metformina/toxicidad , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-jun/genética , Factor de Transcripción Sp1/genética , Activación Transcripcional/efectos de los fármacos
11.
J Exp Clin Cancer Res ; 34: 99, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26362062

RESUMEN

BACKGROUND: Ursolic acid (UA), a pentacyclic triterpenoid, is known to have anti-tumor activity in various cancers including human non small cell lung cancer (NSCLC). However, the molecular mechanisms underlying the action of UA remain largely unknown. METHODS: Cell viability was measured by MTT assays. Apoptosis was analyzed with Annexin V-FITC/PI Apoptosis Detection Kit by Flow cytometry. Western blot analysis was performed to measure the phosphorylation and protein expression of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), DNMT1 [DNA (cytosine-5)-methyltransferase 1], enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and SP1. Exogenous expression of SP1 and DNMT1 was carried out by transient transfection assays. RESULTS: We showed that UA inhibited the growth and induced apoptosis of NSCLC cells in the dose- and time-dependent fashion. Furthermore, we found that UA induced phosphorylation of SAPK/JNK and suppressed the protein expression of DNMT1 and EZH2. The inhibitor of SAPK/JNK (SP600125) blocked the UA-reduced expression of DNMT1 and EZH2. In addition, UA suppressed the expression of SP1 protein. Conversely, overexpression of SP1 reversed the effect of UA on DNMT1 and EZH2 expression, and feedback attenuated UA-induced phosphorylation of SAPK/JNK. Moreover, exogenous expression of DNMT1 antagonized the effect of UA on SAPK/JNK signaling, EZH2 protein expression, and NSCLC cell growth. CONCLUSION: Our results show that UA inhibits growth of NSCLC cells through SAPK/JNK-mediated inhibition of SP1; this in turn results in inhibition the expression of DNMT1 and EZH2. Overexpression of DNMT1 diminishes UA-reduced EZH2 protein expression. The negative feedback regulation of SAPK/JNK signaling by SP1 and DNMT1, and the reciprocal interaction of EZH2 and DNMT1 contribute to the overall effects of UA. This study leads to important new insights into the mechanisms by which UA controls growth of NSCLC cells.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Complejo Represivo Polycomb 2/metabolismo , Triterpenos/farmacología , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1 , Ensayos de Selección de Medicamentos Antitumorales , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal , Ácido Ursólico
12.
J Exp Clin Cancer Res ; 34: 41, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25948105

RESUMEN

BACKGROUND: Baicalein, a natural flavonoid obtained from the Scutellaria baicalensis root, has been reported to inhibit growth of human lung cancer. However, the detailed mechanism underlying this has not been well elucidated. METHODS: Cell viability was measured using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assays. Apoptosis was detected by flow cytometry analysis and caspase 3/7 assays. The expression of RUNX3 and FOXO3a mRNA were measured by real time RT-PCR methods. Western blot analysis was performed to measure the phosphorylation and protein expression of AMP-activated protein kinase alpha (AMPKα) and extracellular signal-regulated kinase 1/2 (ERK1/2), runt-related transcription factor 3 (RUNX3) and forkhead box O3a (FOXO3a). Silencing of FOXO3a and RUNX3 were performed by small interfering RNA (siRNA) methods. Exogenous expression of FOXO3a or RUNX3 was carried out by electroporated transfection assays. RESULTS: We showed that baicalein significantly inhibited growth and induced apoptosis of non-small cell lung cancer (NSCLC) cells in a time- and dose-dependent manner. Baicalein induced RUNX3 and FOXO3a protein expression, and increased phosphorylation of AMPKα and ERK1/2. Moreover, the inhibitors of AMPK and MEK/ERK1/2 reversed the effect of baicalein on RUNX3 and FOXO3a protein expression. Interestingly, while compound C had little effect on blockade of baicalein-induced phosphorylation of ERK1/2, PD98059 significantly abrogated baicalein-induced phosphorylation of AMPKα. Intriguingly, while silencing of RUNX3 abolished the effect of baicalein on expression of FOXO3a and apoptosis, silencing of FOXO3a significantly attenuated baicalein-reduced cell proliferation. On the contrary, overexpression of FOXO3a restored the effect of baicalein on cell growth inhibition in cells silencing of endogenous FOXO3a gene and enhanced the effect of baicalein on RUNX3 protein expression. Finally, exogenous expression of RUNX3 increased FOXO3a protein and strengthened baicalein-induced phosphorylation of ERK1/2. CONCLUSION: Collectively, our results show that baicalein inhibits growth and induces apoptosis of NSCLC cells through AMPKα- and MEK/ERK1/2-mediated increase and interaction of FOXO3a and RUNX3 protein. The crosstalk between AMPKα and MEK/ERK1/2 signaling pathways, and the reciprocal interplay of FOXO3a and RUNX3 converge on the overall response of baicalein. This study reveals a novel mechanism for regulating FOXO3a and RUNX3 signaling axis in response to baicalein and suggests a new strategy for NSCLC associated targeted therapy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Flavanonas/farmacología , Factores de Transcripción Forkhead/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Relación Dosis-Respuesta a Droga , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
13.
Cell Signal ; 26(4): 730-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24412748

RESUMEN

Studies demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) ligands reduce nicotine-induced non small cell lung carcinoma (NSCLC) cell growth through inhibition of nicotinic acetylcholine receptor (nAChR) mediated signaling pathways. However, the mechanisms by which PPARγ ligands inhibited nAChR expression remain elucidated. Here, we show that GW1929, a synthetic PPARγ ligand, not only inhibited but also antagonized the stimulatory effect of acetylcholine on NSCLC cell proliferation. Interestingly, GW1929 inhibited α7 nAChR expression, which was not blocked by GW9662, an antagonist of PPARγ, or by PPARγ siRNA, but was abrogated by the p38 MPAK inhibitor SB239063. GW1929 reduced the promoter activity of α7 nAChR and induced early growth response-1 (Egr-1) protein expression, which was overcame by SB239063, but enhanced by inhibitors of PI3-K and mTOR. Silencing of Egr-1 blocked, while overexpression of Egr-1 enhanced, the effect of GW1929 on α7 nAChR expression and promoter activity. Finally, GW1929 induced Egr-1 bound to specific DNA areas in the α7 nAChR gene promoter. Collectively, these results demonstrate that GW1929 not only inhibits but also antagonizes Ach-induced NSCLC cell growth by inhibition of α7 nAChR expression through PPARγ-independent signals that are associated with activation of p38 MPAK and inactivation of PI3-K/mTOR, followed by inducing Egr-1 protein and Egr-1 binding activity in the α7 nAChR gene promoter. By downregulation of the α7 nAchR, GW1929 blocks cholinergic signaling and inhibits NSCLC cell growth.


Asunto(s)
Benzofenonas/farmacología , Proliferación Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , PPAR gamma/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tirosina/análogos & derivados , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Anilidas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/antagonistas & inhibidores , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Activación Enzimática/efectos de los fármacos , Humanos , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Tirosina/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
Int J Oncol ; 45(1): 95-103, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24806432

RESUMEN

Curcumin, one of the main bioactive components extracted from a traditional Chinese medicinal herb, exhibits potent anticancer activity against many types of cancer cells including nasopharyngeal carcinoma (NPC). However, the detailed molecular mechanism underlying this is not clearly understood. In this study, we showed that curcumin significantly inhibited the growth of NPC cells in a dose- and time-dependent manner as determined by MTT assays, while increasing apoptosis was also observed as measured by flow cytometry for the FITC-Annexin V and propidium iodide (PI) label and Hoechst 33258 staining. To further explore the potential mechanism, we showed that curcumin increased the phosphorylation of ERK1/2 but not p38 MAPK in a time-dependent manner, and induced protein expression of the tumor suppressors FOXO3a and p53 in a dose­dependent manner, which were not observed in the presence of PD98059, an inhibitor of ERK1/2. Furthermore, silencing of FOXO3a and p53 genes by siRNAs overcame the inhibitory effect of curcumin on cell proliferation. Silencing or blockade of p53 using siRNA or chemical inhibitor abrogated the effect of curcumin on expression of FOXO3a protein; silencing or overexpression of FOXO3a had no further effect on curcumin-induced p53 protein expression. Furthermore, blockade of ERK1/2 and exogenous expression of FOXO3a restored the effect of curcumin on growth of cells. Together, our studies show that curcumin inhibits growth and induces apoptosis of NPC cells through ERK1/2-mediated increase in the protein expression and interaction of p53 and FOXO3a. p53 is upstream of FOXO3a, which form a regulatory loop that mediates the effect of curcumin. This study unveils a new mechanism by which curcumin inhibits the proliferation and induces apoptosis of human NPC cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Curcumina/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Nasofaríngeas/patología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Relación Dosis-Respuesta a Droga , Flavonoides/administración & dosificación , Flavonoides/farmacología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Fosforilación , Proteína p53 Supresora de Tumor/metabolismo
15.
J Exp Clin Cancer Res ; 33: 36, 2014 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-24766860

RESUMEN

BACKGROUND: Berberine (BBR), a component from traditional Chinese medicine, has been shown to possess anti-tumor activity against a wide spectrum of cancer cells including human lung cancer, but the detailed mechanism underlining this has not been well elucidated. METHODS: In this study, the effect of berberine on cell growth and apoptosis were assessed by MTT, flow cytometry and Hoechst 33258 staining assays. The phosphorylation of p38 MAPK and ERK1/2, and expressions of p38 MAPK isoforms α and ß, total ERK1/2, p53, FOXO3a and p21 protein were evaluated by Western Blot analysis. Silencing of p38 MAPK isoform α and ß, p53, FOXO3a and p21 were performed by siRNA methods. Exogenous expression of FOXO3a was carried out by electroporated transfection assays. RESULTS: We showed that BBR significantly inhibited growth and induced cell cycle arrest of non small cell lung cancer (NSCLC) cells in the G0/G1 phase in a dose-dependent manner. Furthermore, we found that BBR increased phosphorylation of p38 MAPK and ERK1/2 in a time-dependent and induced protein expression of tumor suppressor p53 and transcription factor FOXO3a in a dose-dependent fashion. The specific inhibitor of p38 MAPK (SB203580), and silencing of p38α MAPK by small interfering RNAs (siRNAs), but not ERK1/2 inhibitor (PD98059) blocked the stimulatory effects of BBR on protein expression of p53 and FOXO3a. Interestingly, inhibition of p53 using one specific inhibitor (Pifithrin-α) and silencing of p53 using siRNAs overcome the inhibitory effect of BBR on cell growth. Silencing of FOXO3a appeared to attenuate the effect of BBR on p53 expression, cell proliferation and apoptosis. Furthermore, BBR induces the protein expression of cell cycle inhibitor p21 (CIP1/WAF1), which was not observed in cells silencing of p53 or FOXO3α gene. Intriguingly, exogenous expression of FOXO3a enhanced the expression of p21 (CIP1/WAF1) and strengthened BBR-induced apoptosis. CONCLUSION: Our results show that BBR inhibits proliferation and induces apoptosis of NSCLC cells through activation of p38α MAPK signaling pathway, followed by induction of the protein expression of p53 and FOXO3a. The latter contribute to the BBR-increased p21 (CIP1/WAF1) protein expression. The exogenous FOXO3a, interaction and mutually exclusive events of p53 and FOXO3a augment the overall response of BBR.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Berberina/farmacología , Factores de Transcripción Forkhead/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma , Adenocarcinoma del Pulmón , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Sistema de Señalización de MAP Quinasas , Fosforilación , Procesamiento Proteico-Postraduccional , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
16.
Cell Signal ; 26(3): 639-47, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24361375

RESUMEN

Nasopharyngeal carcinoma (NPC) is one of the most common cancers of the head and neck, particularly in Southern China and Southeast Asia with high treatment failure due to the development of local recurrence and distant metastasis. The molecular mechanisms related to the progression of NPC have not been fully understood. In this study, we showed that antidiabetes drugs rosiglitazone and metformin inhibit NPC cell growth through reducing the expression of integrin-linked kinase (ILK). Blockade of PPARγ and AMPKα overcame the effects of rosiglitazone and metformin on ILK protein. Importantly, overexpression of ILK abrogated the effect of rosiglitazone and metformin on NPC cell growth. Furthermore, these agents reduced ILK promoter activity, which was not observed in AP-2α, but not Sp1 site mutation in ILK gene promoter. In addition, silencing of AP-2α or overexpression of Sp1 reversed the effect of these agents on ILK protein expression and cell growth. Chromatin immunoprecipitation (ChIP) assay showed that rosiglitazone induced AP-2α, while metformin reduced Sp1 protein binding to the DNA sequences in the ILK gene promoter. Intriguingly, overexpression of Sp1 abolished the effect of rosiglitazone on AP-2α protein expression. Collectively, we show that rosiglitazone and metformin inhibit ILK gene expression through PPARγ- and AMPKα-dependent signaling pathways that are involved in the regulation of AP-2α and Sp1 protein expressions. The effect of combination of rosiglitazone and metformin demonstrates greater extent than single agent alone. The cross-talk of PPARγ and AMPKα signaling enhances the synergistic effects of rosiglitazone and metformin. This study unveils novel mechanisms by which oral antidiabetes drugs inhibit the growth of human NPC cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , PPAR gamma/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Factor de Transcripción Sp1/genética , Factor de Transcripción AP-2/genética , Carcinoma , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoglucemiantes/farmacología , Metformina/farmacología , Mutación , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Regiones Promotoras Genéticas/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Rosiglitazona , Transducción de Señal/genética , Factor de Transcripción Sp1/biosíntesis , Tiazolidinedionas/farmacología , Factor de Transcripción AP-2/biosíntesis
17.
J Exp Clin Cancer Res ; 32: 43, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23867003

RESUMEN

BACKGROUND: N-Acetyl-Cysteine (NAC), a natural sulfur-containing amino acid derivative, and peroxisome proliferators activated receptor alpha (PPARα) ligand have been shown to have anticancer properties. However, the mechanisms by which these agents inhibit human non-small cell lung carcinoma (NSCLC) cell growth have not been well elucidated. METHODS: Small interfering RNAs (siRNAs) were used to knockdown 3-phosphoinositide-dependent protein kinase 1 (PDK1), PPARα, p65 and p53 genes; Western Blot was performed to detect the protein expression of PDK1, PPARα, p65 and p53; Cell viability and MTT assays were carried out to determine the cell proliferation; Transient transfection and Dual-Luciferase Reporter assays were used to transfect siRNAs or exogenous expression vectors, and to measure the gene promoter activity. RESULTS: We showed that NAC inhibited NSCLC cell proliferation through reduction of PDK1 expression. NAC also induced the protein expression of PPARα. While PPARα ligand enhanced, PPARα antagonist and siRNA abrogated the effect of NAC on PDK1 promoter activity, protein expression and cell growth. Overexpression of PDK1 diminished the inhibitory effect of NAC on cell proliferation. NAC induced p53 and reduced p65 protein expression through activation of PPARα. Silencing of p53 and overexpression of p65 blocked the effect of NAC on PDK1 promoter activity and protein expression. CONCLUSION: Our results show that NAC inhibits PDK1 expression through PPARα-mediated induction of p53 and inhibition of p65 protein expression. PPARα ligand enhances the effect of NAC. This ultimately inhibits NSCLC cell growth. This study unveils a novel mechanism by which NAC in combination with PPARα ligand inhibits growth of human lung carcinoma cells.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/antagonistas & inhibidores , Acetilcisteína/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , PPAR alfa/metabolismo , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Silenciador del Gen , Humanos , Neoplasias Pulmonares/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Factor de Transcripción ReIA/genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
18.
Cell Signal ; 25(12): 2566-74, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23998949

RESUMEN

Head and neck cancer is one of the most morbid human malignancies with an overall poor prognosis and severely compromised quality of life. As a result, there is significant interest in developing adjuvant therapies to augment currently available treatment protocols. Curcumin has been found to possess anti-cancer activities via its effect on a variety of biological pathways. In this study, we showed that curcumin inhibits head and neck cancer cell growth through reduction of PGE2 receptor EP4 gene expression. Blockade of AMP-dependent kinase (AMPK), and p38 MAPK by either chemical inhibitors or siRNAs antagonized the inhibitory effect of curcumin on EP4 expression, which was reversed by metformin, an activator of AMPK. Curcumin induced PGC-1α protein that was blocked by compound C and SB239063. Silencing of PGC-1α reversed the effect of curcumin on EP4 protein. Overexpression of EP4 overcame the effect of curcumin on head and neck cancer cell growth. In addition, curcumin reduced Sp1 protein. Overexpression of Sp1 resisted the inhibitory effect of curcumin on EP4 promoter activity and protein expression. Interestingly, overexpression of PGC-1α further enhanced the inhibitory effect of curcumin on Sp1 protein expression that was blocked by SB239063. In conclusion, this study shows that curcumin inhibits EP4 gene expression dependent of AMPKα and p38 MAPK activation, this leads to reduction of Sp1 protein and binding to specific area in the EP4 gene promoter. The cross talks of AMPKα and p38 MAPK signaling, the kinase-mediated PGC-1α expression and reciprocity of PGC-1α and Sp1 enhance this process. This ultimately results in inhibition of head and neck cancer cell proliferation.


Asunto(s)
Antineoplásicos/farmacología , Curcumina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Subtipo EP4 de Receptores de Prostaglandina E/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenilato Quinasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Metformina/farmacología , Terapia Molecular Dirigida , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA