Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Sci Food Agric ; 99(6): 3045-3056, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30488458

RESUMEN

BACKGROUND: This study investigated the in vivo prophylactic effect of probiotic chocolate on constipation. Rats were administered chocolate containing 2.5 × 1010 CFU g-1 of probiotics daily for 4 weeks and treated with loperamide (5 mg kg-1 ) daily at the fourth week of treatment. RESULTS: Probiotic chocolate treatment significantly (P < 0.05) increased the intestinal motility, colon length, fecal moisture content and number of excreted fecal pellets in constipated rats. Moreover, quantitative real-time polymerase chain reaction data and histological images also revealed that both probiotic chocolate LYC and BB12 treatments were capable of upregulating the mRNA expression levels of colonic ZO-1, occludin and AQP8, leading to the maintenance of the defensive barrier function in the constipated rats compared with the negative controls. Interestingly, these treatments also modulated gut bacterial populations by increasing the abundance levels of Lactobacillus and Bifidobacterium, as well as reducing the abundance level of Enterobacteriaceae. CONCLUSION: The present study demonstrated that probiotic chocolate LYC and BB12 could potentially be used as alternative agents for prophylactic constipation. © 2018 Society of Chemical Industry.


Asunto(s)
Chocolate/microbiología , Estreñimiento/prevención & control , Intestinos/fisiopatología , Probióticos/administración & dosificación , Animales , Bifidobacterium animalis/química , Bifidobacterium animalis/metabolismo , Chocolate/análisis , Estreñimiento/fisiopatología , Defecación/efectos de los fármacos , Heces/microbiología , Femenino , Humanos , Lactobacillus plantarum/química , Lactobacillus plantarum/fisiología , Probióticos/química , Ratas , Ratas Sprague-Dawley , Streptococcus thermophilus/química , Streptococcus thermophilus/fisiología
2.
Oncol Lett ; 14(4): 5027-5033, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29085516

RESUMEN

Ginkgetin is a natural biflavonoid isolated from the leaves of Ginkgo biloba, and is characterized by its anti-inflammatory and anti-viral activities. Although numerous studies state that it has also antitumor activity, the anti-proliferative effect of ginkgetin and the underlying mechanism in breast cancer cells have not yet been investigated. In the present study, ginkgetin inhibited the cell viability of MCF-7 and T-47D cells dose-dependently, and suppressed the expression of the estrogen receptor (ER) at the mRNA and protein levels. Among the targets of the ER, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), cyclin D1 and survivin were also downregulated by ginkgetin treatment. The anti-proliferative effects of ginkgetin were sufficient to suppress the growth by estradiol stimulation. However, ginkgetin did not significantly affect the viability of MDA-MB-231 cells, which are ER-negative cells. Furthermore, the knockdown of the ER and an inhibitor of PFKFB3 significantly sensitized MCF-7 and T-47D cells to ginkgetin. These findings suggest that ginkgetin induces cell death in ER-positive breast cancer cells via the inhibition of ER expression and that it is a promising agent for breast cancer treatment.

3.
Oncotarget ; 7(37): 59809-59819, 2016 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-27494858

RESUMEN

Metabolic reprogramming in cancer cells has recently been recognized as an essential hallmark of neoplasia. In this context, metabolic alterations represent an attractive therapeutic target, and encouraging results with drugs targeting various metabolic processes have been obtained in preclinical studies. Recently, several studies have suggested that dichloroacetate (DCA), a specific pyruvate dehydrogenase kinase inhibitor, may be a potential anticancer drug in a large number of diverse tumors. However, the precise mechanism is not fully understood, which is important for the use of DCA in cancer treatment. In the present study, we found that DCA sensitized MCF7 breast cancer cells to tamoxifen-induced cell death by decreasing epidermal growth factor receptor (EGFR) expression. The downregulation of EGFR was caused by degradation of the protein. Furthermore, p38 mitogen-activated protein kinase played an important role in DCA/tamoxifen-induced EGFR degradation. Finally, DCA also promoted comparable tamoxifen-induced cell death in tamoxifen-resistant MCF7 cells, which were established by long-term treatment with tamoxifen. In summary, our results suggest that DCA is an attractive potential drug that sensitizes cells to tamoxifen-induced cell death and overcome tamoxifen resistance via downregulation of EGFR expression in breast cancer cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ácido Dicloroacético/farmacología , Receptores ErbB/metabolismo , Muerte Celular/efectos de los fármacos , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Proteolisis/efectos de los fármacos , Transducción de Señal , Tamoxifeno/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Oncotarget ; 6(28): 26528-40, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25605253

RESUMEN

In the present study, we found that selective inhibition of histone deacetylase 2 (HDAC2) with small inhibitory RNA (siRNA) induced survivin downregulation in a p53-dependent manner. Interestingly, suberoylanilide hydroxamic acid (SAHA) or knockdown of HDAC2 induced downregulation of Mdm2, a negative regulator of p53, at the protein level. SAHA and/or HDAC2 siRNA increased Mdm2 ubiquitination, and MG132, an inhibitor of proteosome function, prevented HDAC2 inhibition-induced degradation of Mdm2. Clinically, the mRNA levels of HDAC2 and survivin were prominently overexpressed in lung cancer patients compared to normal lung tissues. Silencing of HDAC2 enhanced the cell death caused by ionizing radiation in lung cancer cells. Collectively, our results indicate that selective inhibition of HDAC2 causes survivin downregulation through activation of p53, which is mediated by downregulation of Mdm2. They further suggest that HDAC2 may exert a dominant effect on lung cancer cell survival by sustaining Mdm2-survivin levels.


Asunto(s)
Histona Desacetilasa 2/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Regulación hacia Abajo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma/farmacología , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , Survivin , Transfección , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
5.
Biochem Biophys Res Commun ; 453(3): 438-42, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25281537

RESUMEN

Cyclin A1 is an A-type cyclin that directly binds to CDK2 to regulate cell-cycle progression. In the present study, we found that doxorubicin decreased the expression of cyclin A1 at the protein level in A549 lung cancer cells, while markedly downregulating its mRNA levels. Interestingly, doxorubicin upregulated caspase-1 in a concentration-dependent manner, and z-YAVD-fmk, a specific inhibitor of caspase-1, reversed the doxorubicin-induced decrease in cyclin A1 in A549 lung cancer and MCF7 breast cancer cells. Active caspase-1 effectively cleaved cyclin A1 at D165 into two fragments, which in vitro cleavage assays showed were further cleaved by caspase-3. Finally, we found that overexpression of cyclin A1 significantly reduced the cytotoxicity of doxorubicin, and knockdown of cyclin A1 by RNA interference enhanced the sensitivity of cells to ionizing radiation. Our data suggest a new mechanism for the downregulation of cyclin A1 by DNA-damaging stimuli that could be intimately involved in the cell death induced by DNA damage-inducing stimuli, including doxorubicin and ionizing radiation.


Asunto(s)
Caspasas/metabolismo , Muerte Celular/genética , Ciclina A1/metabolismo , Daño del ADN , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Humanos , Proteolisis , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Biochem Biophys Res Commun ; 449(4): 490-5, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24845567

RESUMEN

TWIST1 is implicated in the process of epithelial mesenchymal transition, metastasis, stemness, and drug resistance in cancer cells, and therefore is a potential target for cancer therapy. In the present study, we found that knockdown of TWIST1 by small interfering RNA (siRNA) enhanced arsenic trioxide (ATO)- and ionizing radiation (IR)-induced cell death in non-small-cell lung cancer cells. Interestingly, intracellular reactive oxygen species levels were increased in cells treated with TWIST1 siRNA and further increased by co-treatment with ATO or IR. Pretreatment of lung cancer cells with the antioxidant N-acetyl-cysteine markedly suppressed the cell death induced by combined treatment with TWIST1 siRNA and ATO or IR. Moreover, treatment of cells with TWIST1 siRNA induced mitochondrial membrane depolarization and significantly increased mitochondrial fragmentation (fission) and upregulated the fission-related proteins FIS1 and DRP1. Collectively, our results demonstrate that siRNA-mediated TWIST1 knockdown induces mitochondrial dysfunction and enhances IR- and ATO-induced cell death in lung cancer cells.


Asunto(s)
Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Acetilcisteína/farmacología , Trióxido de Arsénico , Arsenicales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Técnicas de Silenciamiento del Gen , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Óxidos , ARN Interferente Pequeño/farmacología , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis
7.
J Thorac Oncol ; 6(8): 1313-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21642861

RESUMEN

INTRODUCTION: Histone deacetylase inhibitors (HDACIs) induce growth arrest and apoptosis in cancer cells. In addition to their intrinsic anticancer properties, HDACIs modulate cellular responses to ionizing radiation (IR). We examined the molecular mechanism(s) associated with the radiosensitizing effects of HDACIs in human lung cancer cells. METHODS: Lung cancer cells were pretreated with the appropriate concentrations of suberoylanilide hydroxamic acid or trichostatin A. After 2 hours, cells were irradiated with various doses of γ-IR, and then we performed 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, fluorescence-activated cell sorting analysis, clonogenic assay, and Western blotting to detect cell viability or apoptosis and changes of specific proteins expression levels. RESULTS: In this study, we showed that HDACIs (including suberoylanilide hydroxamic acid and trichostatin A) and IR synergistically trigger cell death in human non-small cell lung cancer cells. Cell viability and clonogenic survival were markedly decreased in cultures cotreated with HDACIs and IR. Interestingly, p53 acetylation at lysine 382 was significantly increased, and c-myc expression simultaneously down-regulated in cotreated cells. Radiosensitization by HDACIs was inhibited on transfection with small interfering RNA against p53 and c-myc overexpression, supporting the involvement of p53 and c-myc in this process. Furthermore, c-myc down-regulation and apoptotic cell death coinduced by IR and HDACI were suppressed in cells transfected with mutant K382R p53 and C135Y p53 displaying loss of acetylation at lysine 382 and DNA-binding activity, respectively. CONCLUSIONS: Our results collectively demonstrate that the degree of radiosensitization by HDACIs is influenced by acetyl p53-mediated c-myc down-regulation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pulmonares/terapia , Proteínas Proto-Oncogénicas c-myc/metabolismo , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Acetilación/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Regulación hacia Abajo , Citometría de Flujo , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de la radiación , Mutagénesis Sitio-Dirigida , Mutación/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteína p53 Supresora de Tumor/genética , Vorinostat
8.
Biochem Pharmacol ; 82(3): 216-26, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21601561

RESUMEN

Sorafenib, a multikinase inhibitor, is emerging as a promising targeted agent that may possess antitumor activity against a broad range of cancers. The mechanism by which sorafenib induces lung cancer cell death and apoptosis, however, is not understood. In the present study, we provide evidence that sorafenib acts through inhibition of mammalian target of rapamycin (mTOR) to down-regulate survivin and promote apoptotic cell death in human non-small cell lung cancer (NSCLC) cells. Sorafenib induced ATF4-mediated Redd1 expression, leading to mTOR inhibition-the upstream signal for down-regulation of survivin. Overexpression of survivin reduced sorafenib-induced apoptosis, whereas silencing survivin using small interfering RNA (siRNA) enhanced it, supporting the interpretation that down-regulation of survivin is involved in sorafenib-induced cell death in human NSCLC cells. Furthermore, sorafenib abolished the induction of survivin that normally accompanies IGF-1-stimulated mTOR activation. We further found that Redd1-induced mTOR down-regulation and ATF4/CHOP-induced expression of the TRAIL receptor DR5 associated with sorafenib treatment helped sensitize cells to TRAIL-induced apoptosis. Our study suggests that sorafenib mediates apoptotic cell death in human NSCLC cells through Redd1-induced inhibition of mTOR and subsequent down-regulation of survivin, events that are associated with sensitization to TRAIL-induced apoptotic cell death.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bencenosulfonatos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Piridinas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Sorafenib , Survivin , Serina-Treonina Quinasas TOR/genética
9.
Biochem Biophys Res Commun ; 407(3): 507-11, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21414293

RESUMEN

Redd1 acts as a negative regulator of mTOR in response to various stress conditions, but its specific physiological role is currently unclear. In the present study, we showed that Redd1 inhibits the invasive activity of non-small cell lung cancer (NSCLC) cells. Interestingly, expression of Redd1 was extremely low in H1299 cells displaying high invasiveness, compared with that in H460 cells with lower invasive activity. Overexpression of Redd1 inhibited the invasive activity of H1299 cells, while suppression with specific siRNAs enhanced the invasiveness of H460 cells. Knockdown of the mTOR downstream substrate, S6K, resulted in a decrease in the invasive property of H1299 cells. Our results provide preliminary evidence that Redd1 inhibits the invasive activity of NSCLC cells via suppression of the mTOR downstream pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factores de Transcripción/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Células HeLa , Humanos , Neoplasias Pulmonares/metabolismo , Invasividad Neoplásica , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/genética
10.
IUBMB Life ; 61(12): 1153-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19946894

RESUMEN

Nuclear protein 1 (NUPR1) was originally identified as p8, a member of the family of HMG-I/Y transcription factors induced in response to various cellular stressors. However, the signaling pathway underlying NUPR1 induction by cellular stresses remains to be established. In this study, we found that the expression of NUPR1 by various stresses induced by activating transcription factor 4 (ATF4). Loss of ATF4 using siRNA significantly diminished NUPR1 expression. Overexpression of ATF4 caused NUPR1 levels to rise. NUPR1 expression was associated with enhanced transcriptional activation of genes of ATF4 downstream, suggesting that the protein promoted the transcription of stress-regulated genes via positive feedback on the ATF4 pathway.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Activación Transcripcional , Línea Celular Tumoral , Retroalimentación Fisiológica , Células HeLa , Humanos , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba
11.
J Biol Chem ; 284(45): 30871-80, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19740745

RESUMEN

The Hsp90-associated protein p23 modulates Hsp90 activity during the final stages of the chaperone pathway to facilitate maturation of client proteins. Previous reports indicate that p23 cleavage induced by caspases during cell death triggers destabilization of client proteins. However, the specific role of truncated p23 (Delta p23) in this process and the underlying mechanisms remain to be determined. One such client protein, hTERT, is a telomerase catalytic subunit regulated by several chaperone proteins, including Hsp90 and p23. In the present study, we examined the effects of p23 cleavage on hTERT stability and telomerase activity. Our data showed that overexpression of Delta p23 resulted in a decrease in hTERT levels, and a down-regulation in telomerase activity. Serine phosphorylation of Hsp90 was significantly reduced in cells expressing high levels of Delta p23 compared with those expressing full-length p23. Mutation analyses revealed that two serine residues (Ser-231 and Ser-263) in Hsp90 are important for activation of telomerase, and down-regulation of telomerase activity by Delta p23 was associated with inhibition of cell growth and sensitization of cells to cisplatin. Our data aid in determining the mechanism underlying the regulation of telomerase activity by the chaperone complex during caspase-dependent cell death.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Proteínas HSP90 de Choque Térmico/metabolismo , Telomerasa/metabolismo , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas HSP90 de Choque Térmico/genética , Humanos , Fosforilación , Estabilidad Proteica , Telomerasa/genética
12.
Int J Oncol ; 35(1): 81-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19513554

RESUMEN

Ionizing radiation (IR) is widely used in cancer treatment and in biological studies. It disrupts cellular homeostasis through multiple mechanisms including changes of the expression profile of genes. Although microRNAs (miRNAs) have recently been recognized as important post-transcriptional regulators and are involved in various biological processes, whether miRNAs play any roles in the cellular response to IR, is not well examined. We investigated the profile of miRNA expression following IR in the human lung carcinoma cell line A549, and the expression profiles of IR-responsive miRNAs were confirmed by qRT-PCR. The target mRNAs of IR-responsive miRNAs were predicted with a target prediction tool. Microarray analysis identified 12 and 18 miRNAs in 20- and 40 Gy-exposed A549 cells, respectively, that exhibited more than 2-fold changes in their expression levels. Of these, four were changed in only 20-Gy-treated cells, ten only in 40-Gy-treated cells, and eight miRNAs were found to change after both treatments. qRT-PCR analysis of a subset of the miRNAs showed patterns of regulation as the microarray data, although the magnitude of the changes differed in the two data sets. Target prediction for IR-responsive miRNAs suggests that they target genes related to apoptosis, regulation of cell cycle, and DNA damage and repair. Taken together, these data suggest that miRNA expression is affected by radiation, and they may be involved in the regulation of radiation responses.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Neoplasias Pulmonares/genética , MicroARNs/efectos de la radiación , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Perfilación de la Expresión Génica/métodos , Humanos , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Free Radic Biol Med ; 46(8): 1158-67, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19439225

RESUMEN

Regulation of mRNA translation in mammalian cells involves the coordinated control of mammalian target of rapamycin (mTOR) signaling. At present, limited information is available on the potential relevance of mTOR regulation, although translation inhibition during oxidative and endoplasmic reticulum (ER) stress is clearly important. In this study, we show that activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein-beta (C/EBP-beta) negatively regulate mTOR via Redd1 expression in response to oxidative and ER stress. Oxidative and ER stress conditions induce rapid and significant activation of ATF4 downstream of eIF2alpha phosphorylation, which is responsible for Redd1 expression. In our experiment, overexpression of ATF4 was associated with reduced mTOR activity via Redd1 expression, whereas suppression of ATF4 levels with small interfering RNA led to the recovery of decreased mTOR activity mediated by downregulation of Redd1 during oxidative and ER stress. We additionally identified Redd1 as a downstream effector of C/EBP-beta stimulated by ATF4 activated under the stress conditions examined. RNA interference studies provided further evidence of the requirement of C/EBP-beta for Redd1 expression. We conclude that the Redd1 gene is transactivated by the ATF4 and C/EBP family of transcription factors, leading to mTOR inhibition in response to oxidative and ER stress.


Asunto(s)
Factor de Transcripción Activador 4/fisiología , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Factores de Transcripción/metabolismo , Activación Transcripcional , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Retículo Endoplásmico , Retroalimentación Fisiológica , Células HeLa , Humanos , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Sirolimus/metabolismo , Serina-Treonina Quinasas TOR , Factores de Transcripción/genética
14.
FEBS Lett ; 583(1): 123-7, 2009 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19059405

RESUMEN

SP600125 (SAPK Inhibitor II) is reported to function as a reversible ATP competitive inhibitor of c-Jun N-terminal kinase (JNK). In the present study, we show that SP600125 induces a dose-dependent decrease in mTOR activity, as assessed by reduced phosphorylation of the downstream targets S6K1 and S6, and a significant increase in the expression of Redd1. Knockdown of Redd1 expression by siRNA resulted in a recovery of decreased S6 phosphorylation by SP600125. Overexpression of ATF4 upregulated the expression of Redd1, while suppression of ATF4 expression by siRNA enhanced the level of S6 phosphorylation by downregulating the SP600125-induced increase in Redd1 expression. Together, these results indicate that SP600125 inhibits mTOR activity via an ATF4-induced increase in Redd1 expression.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Antracenos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Sirolimus/farmacología , Factores de Transcripción/biosíntesis , Factor de Transcripción Activador 4/genética , Células HeLa , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas/metabolismo , ARN Interferente Pequeño/genética , Serina-Treonina Quinasas TOR
15.
Mol Cancer Res ; 6(6): 996-1002, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18567803

RESUMEN

Epidermal growth factor receptor (EGFR) is activated by ionizing radiation (IR), but the molecular mechanism for this effect is unknown. We have found that intracellular generation of nitric oxide (NO) by NO synthase (NOS) is required for the rapid activation of EGFR phosphorylation by IR. Treatment of A549 lung cancer cells with IR increased NOS activity within minutes, accompanied by an increase of NO. 2-Phenyl-4,4,5,5,-tetramethylimidazolline-1-oxyl-3-oxide, an NO scavenger, and NG-monomethyl-l-arginine, an NOS inhibitor, abolished the increase in intracellular NO and activation of EGFR by IR. In addition, an NO donor alone induced EGFR phosphorylation. Transient transfection with small interfering RNA for endothelial NOS reduced IR-induced NO production and suppressed IR-induced EGFR activation. Overexpression of endothelial NOS increased IR-induced NO generation and EGFR activation. These results indicate a novel molecular mechanism for EGFR activation by IR-induced NO production via NOS.


Asunto(s)
Receptores ErbB/metabolismo , Óxido Nítrico/biosíntesis , Radiación Ionizante , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal
16.
Oncol Rep ; 19(6): 1577-82, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18497968

RESUMEN

A-kinase-anchoring protein 149 (AKAP149) is a member of a structurally diverse, though functionally similar anchoring protein family and is localized to the outer membrane of mitochondria and in the endoplasmic reticulum-nuclear envelope network. AKAP149 plays an important role in controlling the subcellular localization and temporal specificity of protein phosphorylation and mRNA metabolism by tethering kinases and phosphatases, such as protein kinase A and type I protein phosphatase, through its N-terminal protein-binding motifs and mRNAs via its C-terminal RNA-binding motifs. It is well recognized that caspases play a central role in transducing and amplifying the intracellular death signal and that apoptosis is executed as a consequence of caspase-mediated cleavage of multiple cellular substrates. The identification of novel death substrates and elucidation of the consequences of their proteolytic cleavages by caspases are therefore crucial for our understanding of cell death and other biological processes. Herein, we demonstrated that AKAP149 is a direct substrate of active caspase-3, -8 -and -10 in vitro and in vivo. 35S-labeled full-length AKAP149 was completely cleaved in vitro by active caspase-3, -8 and -10 into two fragments of approximately 105 and 45 kDa, while caspase-2 cleaved it partially and caspase-1 did not cleave it at all. AKAP149 was also cleaved by caspases during Fas- and staurosporine-induced apoptosis in Jurkat T and HeLa cells, which were blocked by specific inhibitors of caspase-3 and -8. The specific cleavage site for these caspases was mapped in vitro and in vivo to Asp582 at AKAP149, which is located between the protein kinase A regulatory subunit anchoring and KH RNA-binding domains. In addition, HeLa cells transiently overexpressing AKAP149 D582E mutant were resistant to staurosporine-induced HeLa cell apoptosis. Taken together, these data suggest that AKAP149 activity may be deregulated by caspase-dependent proteolysis during apoptotic cell death and may provide useful information for elucidating the apoptosis signaling pathways in detail.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Apoptosis/fisiología , Caspasas/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Ácido Aspártico/química , Ácido Aspártico/genética , Células HeLa , Humanos , Células Jurkat , Mutación/genética , Estaurosporina/farmacología , Receptor fas/farmacología
17.
Lung Cancer ; 61(3): 317-27, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18281123

RESUMEN

SUMMARY: In the present study, we show that a combination of sulindac and arsenic trioxide (ATO) induces more extensive apoptosis than either drug alone in H1299 human non-small cell lung carcinoma (NSCLC) cells. Treatment with sulindac/ATO triggered three major apoptotic signaling events, namely, collapse of the mitochondrial membrane potential, release of cytochrome c, and activation of caspases. Furthermore, the sulindac/ATO combination induced reactive oxygen species (ROS) generation, and the antioxidant, N-acetyl-L-cysteine, blocked this apoptotic signaling. The c-Jun NH(2)-terminal kinase (JNK) was activated downstream of ROS production in H1299 cells. Blockage of JNK by pretreatment with SP600125, a pharmacological inhibitor, or transfection with dominant-negative (DN) JNK1 vectors abrogated sulindac/ATO-induced apoptosis, as evident from the disruption of caspase activation. Interestingly, a slower migrating Bcl-xL band was observed on immunoblots after treatment of cells with sulindac/ATO. The band was absent upon the treatment of cell lysates with lambda protein phosphatase. Moreover, confocal microscopy findings disclose that active JNK translocates to mitochondria. Treatment with SP600125 and transfection with DN-JNK blocked Bcl-xL phosphorylation, suggesting that JNK plays an important role in sulindac/ATO-induced Bcl-xL phosphorylation. In conclusion, in H1299 human NSCLC cells, sulindac and ATO synergistically induce a high degree of apoptosis, which is mediated by the ROS-dependent JNK activation pathway via Bcl-xL phosphorylation.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/patología , Óxidos/farmacología , Sulindac/farmacología , Proteína bcl-X/metabolismo , Acetilcisteína/farmacología , Trióxido de Arsénico , Arsenicales/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral/metabolismo , Sinergismo Farmacológico , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Óxidos/metabolismo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sulindac/metabolismo , Proteína bcl-X/fisiología
18.
Mol Pharmacol ; 73(3): 1005-12, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18156316

RESUMEN

Histone deacetylase (HDAC) inhibitors represent a promising group of anticancer agents. Treatment of cancer cells with HDAC blockers, such as suberoylanilide hydroxamic acid (SAHA), leads to the activation of apoptosis-promoting genes. To enhance proapoptotic efficiency, SAHA has been used in conjunction with radiation, kinase inhibitors, and cytotoxic drugs. In the present study, we show that at the suboptimal dose of 250 muM, sulindac [2-[6-fluoro-2-methyl-3-[(4-methylsulfinylphenyl)methylidene]inden-1-yl]-acetic acid] significantly enhances SAHA-induced growth suppression and apoptosis of A549 human non-small cell lung cancer cells, primarily via enhanced collapse of the mitochondrial membrane potential, release of cytochrome c, and caspase activation. Furthermore, sulindac/SAHA cotreatment induced marked down-regulation of survivin at both the mRNA and protein levels and stimulated the production of reactive oxygen species (ROS), which were blocked by the antioxidant N-acetyl-l-cysteine. Overexpression of survivin was associated with reduced sulindac/SAHA-induced apoptosis of A549 cells, whereas suppression of survivin levels with antisense oligonucleotides or small interfering RNA further sensitized cells to sulindac/SAHA-induced cell death. Our results collectively demonstrate that sulindac/SAHA-induced apoptosis is mediated by ROS-dependent down-regulation of survivin in lung cancer cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Sulindac/farmacología , Anexina A5/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Caspasas/análisis , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Fluoresceína-5-Isotiocianato/metabolismo , Colorantes Fluorescentes/metabolismo , Inhibidores de Histona Desacetilasas , Humanos , Indicadores y Reactivos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Propidio/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Vorinostat
19.
Cell Signal ; 19(7): 1393-403, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17307335

RESUMEN

Redd1, a recently discovered stress-response gene, is regulated by hypoxia via hypoxia-inducible factor 1 (HIF-1) and by DNA damage via p53/p63; however, the signaling pathway by which its expression is induced by hypoxia has not been elucidated. In the present study, we demonstrated that the expression of Redd1 in response to hypoxia (1% O(2)), hypoxia-mimetic agent, cobalt chloride (CoCl(2)) and high cell density (HCD) requires coactivation of HIF-1alpha and Sp1. CoCl(2) and HCD induced the activation of HIF-1alpha and Sp1 in HeLa cells, and siRNAs targeting HIF-1alpha and Sp1 abrogated Redd1 expression. Inhibition of phosphatidylinositol 3-kinase (PI3K) by LY294002 and by a dominant-negative PI3K mutant reduced the expression of Redd1 and activation of HIF-1alpha and Sp1 by CoCl(2) and HCD. Also, suppression of Akt activation blocked the expression of Redd1 and the activation of HIF-1alpha and Sp1 by CoCl(2) and HCD. Furthermore, we found that the induction of Redd1 expression by CoCl(2) can be mediated by activation of Sp1 in HIF-1alpha-deficient cells but that a higher level of Redd1 expression is achieved when these cells are transfected with HIF-1alpha. These results demonstrate that hypoxic condition-and HCD-induced expression of Redd1 is mediated by coactivation of Sp1 and HIF-1alpha downstream of the PI3K/Akt signaling pathway.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión , Recuento de Células , Hipoxia de la Célula , Secuencia de Consenso , Activación Enzimática , Regulación de la Expresión Génica , Células HeLa , Humanos , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética
20.
Apoptosis ; 12(1): 195-209, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17136320

RESUMEN

Non-steroidal anti-inflammatory drugs are well known to induce apoptosis of cancer cells independent of their ability to inhibit cyclooxygenase-2, but the molecular mechanism for this effect has not yet been fully elucidated. The purpose of this study was to elucidate the potential signaling components underlying sulindac-induced apoptosis in human multiple myeloma (MM) cells. We found that sulindac induces apoptosis by promoting ROS generation, accompanied by opening of mitochondrial permeability transition pores, release of cytochrome c and apoptosis inducing factor from mitochondria, followed by caspase activation. Bcl-2 cleavage and down-regulation of the inhibitor of apoptosis proteins (IAPs) family including cIAP-1/2, XIAP, and survivin, occurred downstream of ROS production during sulindac-induced apoptosis. Forced expression of survivin and Bcl-2 blocked sulindac-induced apoptosis. Most importantly, sulindac-derived ROS activated p38 mitogen-activated protein kinase and p53. SB203580, a p38 mitogen-activated protein kinase inhibitor, and RNA inhibition of p53 inhibited the sulindac-induced apoptosis. Furthermore, p53, Bax, and Bak accumulated in mitochondria during sulindac-induced apoptosis. All of these events were significantly suppressed by SB203580. Our results demonstrate a novel mechanism of sulindac-induced apoptosis in human MM cells, namely, accumulation of p53, Bax, and Bak in mitochondria mediated by p38 MAPK activation downstream of ROS production.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sulindac/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Survivin , Proteína p53 Supresora de Tumor/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA