Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 14: 1312683, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38074160

RESUMEN

[This corrects the article DOI: 10.3389/fphar.2022.907826.].

2.
Front Pharmacol ; 13: 907826, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35721174

RESUMEN

We characterized a so-called "heirloom recipe" Chinese herbal formula (temporarily named Formula X) that contains five Chinese medical botanical drugs, Huang-Lian (Coptis chinensis Franch. [Ranunculaceae]), Huang-Qin (Scutellaria baicalensis Georgi [Lamiaceae]), Bai-Wei (Vincetoxicum atratum (Bunge) C. Morren and Decne. [Apocynaceae]), E-Zhu (Curcuma aromatica Salisb. [Zingiberaceae]) and Bai-Zhu (Atractylodes macrocephala Koidz. [Asteraceae]). Formula X inhibited the growth of various cancer cells and decreased the expression levels of a panel of proteins, including CD133, Myc, PD-L1, and Slug, in cancer cells. We further found that the inhibition of growth and protein expression were exerted by Huang-Lian, Huang-Qin, and Bai-Wei (formula HHB), which exhibited the same biological effects as those of Formula X. Furthermore, we selected three active chemicals, berberine, baicalin, and saponin from Huang-Lian, Huang-Qin, and Bai-Wei, respectively, to produce a chemical formulation (formula BBS), which exhibited similar effects on cell growth and protein expression as those induced by formula HHB. Both the formulae HHB and BBS suppressed tumor growth in an animal study. Moreover, they decreased the protein levels of Myc and PD-L1 in tumor cells in vivo. In summary, we established a novel Chinese herbal formula and a chemical formula that targeted three important processes, tumor maintenance (tumor stem cells), progression, and metastasis, and that influenced the response of tumors to host immunosuppression, for the potentially effective treatment of cancer patients.

3.
Mol Cancer ; 10: 53, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21575199

RESUMEN

BACKGROUND: Both IκB kinase (IKK) complex and oncgenic protein Myc play important roles in cancer progression, including cancer cell invasiveness and metastasis. The levels of Myc is regulated by the phosphorylation of Myc at Thr58 and Ser62. RESULTS: In this study, we show that the expression of Myc is associated with IKKα and IKKß in breast cancers and that Myc is an IKKs substrate. Suppression of IKK activity by either chemical inhibitor or transfection of kinase-dead mutants decreases the phosphorylation of Myc at Ser62 and enhances the degradation of Myc. Consequently, these treatments decrease the tumorigenic and invasive ability of breast cancer cells. Furthermore, doxorubicin, a frequently used anticancer drug in breast cancer, activates IKKs and Myc, thereby increasing invasiveness and tumorigenesis of breast carcinoma MCF7 cells. Inhibition of IKKs prevents these doxorubicin-induced effects. CONCLUSIONS: Our study indicates that IKKs tightly regulate Myc expression through prolonging protein stability, and suggests that IKKs are potentially therapeutic targets and that suppression of IKKs may be used following chemotherapy to reduce the risk of treatment-induced tumor progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Quinasa I-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Doxorrubicina/farmacología , Femenino , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Nitrilos/farmacología , Unión Proteica/fisiología , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sulfonas/farmacología
4.
Eur J Cancer ; 47(8): 1244-57, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21334199

RESUMEN

mTOR (mammalian target of rapamycin) inhibitors were recently found to be effective in the treatment of various human non-Hodgkin's lymphomas (NHLs). We recently reported that RAD001, an mTOR inhibitor, suppressed the growth of lymphoma cells at concentrations much lower than those required for carcinomas. However, the basis for the enhanced sensitivity to RAD001 is unknown. Seven aggressive NHL cell lines and seven carcinoma cell lines were used in this study. Cell cycle was analysed by flow cytometry. pAKT (phosphorylated AKT) (Ser(473) and Thr(308)), p-p70S6K, p-4E-BP1, p-mTOR, p-eIF4E (phosphorylated eIF4E), cyclin A, cyclin E, cyclin D3, c-Myc and insulin receptor substrate-1 (IRS-1) protein expression were assessed by immunoblotting. The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) signalling pathway was constitutively expressed in all seven lymphoma cell lines. RAD001 down-regulated p-mTOR, p-p70S6K, p-4E-BP1, cyclin A, cyclin E, cyclin D3, and c-Myc, but did not affect IRS-1. In parallel with RAD001-induced inhibition of cell viability, a dose- and schedule- dependent down-regulation of pAKT and p-eIF4E expressions was demonstrated. In contrast, a compensatory activation of pAKT and p-eIF4E, was observed in seven carcinoma cells. These findings indicate that the basis for enhanced activity of mTOR inhibitors in NHL may be the lack of compensatory activation of AKT and eIF4E phosphorylation in lymphoma cells.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Regulación Neoplásica de la Expresión Génica , Linfoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/metabolismo , Apoptosis , Carcinoma/metabolismo , Ciclo Celular , Línea Celular Tumoral , Everolimus , Humanos , Inmunosupresores/farmacología , Células Jurkat , Modelos Biológicos , Fosforilación , Sirolimus/farmacología , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
5.
Cancer Res ; 70(22): 9309-18, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21062976

RESUMEN

Markers that could accurately predict responses to the general kinase inhibitor sorafenib are needed to better leverage its clinical applications. In this study, we examined a hypothesized role in the drug response for the growth arrest DNA damage-inducible gene 45ß (GADD45ß), which is commonly underexpressed in hepatocellular carcinoma (HCC) where sorafenib may offer an important new therapeutic option. The anticancer activity of sorafenib-induced GADD45ß expression was tested in a panel of HCC cell lines and xenograft models. We found that GADD45ß mRNA and protein expression were induced relatively more prominently in HCC cells that were biologically sensitive to sorafenib treatment. GADD45ß induction was not found after treatment with either the mitogen-activated protein kinase-extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor U0126 or the Raf inhibitor ZM336372, suggesting that GADD45ß induction by sorafenib was independent of Raf/MEK/ERK signaling activity. However, c-Jun NH2-terminal kinase (JNK) kinase activation occurred preferentially in sorafenib-sensitive cells. Small interfering RNA-mediated knockdown of GADD45ßor JNK kinase limited the proapoptotic effects of sorafenib in sorafenib-sensitive cells. We defined the -339/-267 region in the GADD45ß promoter containing activator protein-1 and SP1-binding sites as a crucial region for GADD45ß induction by sorafenib. Together, our findings suggest that GADD45ß induction contributes to sorafenib-induced apoptosis in HCC cells, prompting further studies to validate its potential value in predicting sorafenib efficacy.


Asunto(s)
Antígenos de Diferenciación/genética , Apoptosis/efectos de los fármacos , Bencenosulfonatos/farmacología , Carcinoma Hepatocelular/genética , Piridinas/farmacología , Animales , Antracenos/farmacología , Antígenos de Diferenciación/metabolismo , Antineoplásicos/farmacología , Sitios de Unión/genética , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sorafenib , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción AP-1/metabolismo , Trasplante Heterólogo
6.
Cancer Lett ; 298(2): 195-203, 2010 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-20673698

RESUMEN

Single-agent mammalian target of rapamycin complex 1 (mTORC1) inhibitors have recently been reported as effective salvage treatment in non-Hodgkin lymphoma (NHL). The combined effect of mTORC1 inhibitor, RAD001, with chemotherapeutic agents used for relapsed or refractory NHL was examined. Synergistic interactions were observed for RAD001 plus gemcitabine or paclitaxel in six NHL cell lines; enhanced gemcitabine- and paclitaxel-induced caspase-dependent apoptosis associated with down-regulation of mTOR signaling was detected. Synergistic interactions were also observed with RAD001 plus gemcitabine and paclitaxel. In conclusion, synergistic cytotoxicity was observed with RAD001 plus gemcitabine and paclitaxel in NHL cells. Combination therapy with these three drugs should be examined in patients with refractory or relapsed NHL.


Asunto(s)
Desoxicitidina/análogos & derivados , Paclitaxel/farmacología , Sirolimus/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Everolimus , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/farmacología , Células Jurkat , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma no Hodgkin/metabolismo , Linfoma no Hodgkin/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Paclitaxel/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Gemcitabina
7.
J Biol Chem ; 284(17): 11121-33, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19261616

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common and aggressive human malignancies. Recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anti-tumor agent. However, many HCC cells show resistance to TRAIL-induced apoptosis. In this study, we showed that bortezomib, a proteasome inhibitor, overcame TRAIL resistance in HCC cells, including Huh-7, Hep3B, and Sk-Hep1. The combination of bortezomib and TRAIL restored the sensitivity of HCC cells to TRAIL-induced apoptosis. Comparing the molecular change in HCC cells treated with these agents, we found that down-regulation of phospho-Akt (P-Akt) played a key role in mediating TRAIL sensitization of bortezomib. The first evidence was that bortezomib down-regulated P-Akt in a dose- and time-dependent manner in TRAIL-treated HCC cells. Second, LY294002, a PI3K inhibitor, also sensitized resistant HCC cells to TRAIL-induced apoptosis. Third, knocking down Akt1 by small interference RNA also enhanced TRAIL-induced apoptosis in Huh-7 cells. Finally, ectopic expression of mutant Akt (constitutive active) in HCC cells abolished TRAIL sensitization effect of bortezomib. Moreover, okadaic acid, a protein phosphatase 2A (PP2A) inhibitor, reversed down-regulation of P-Akt in bortezomib-treated cells, and PP2A knockdown by small interference RNA also reduced apoptosis induced by the combination of TRAIL and bortezomib, indicating that PP2A may be important in mediating the effect of bortezomib on TRAIL sensitization. Together, bortezomib overcame TRAIL resistance at clinically achievable concentrations in hepatocellular carcinoma cells, and this effect is mediated at least partly via inhibition of the PI3K/Akt pathway.


Asunto(s)
Ácidos Borónicos/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Antineoplásicos/farmacología , Apoptosis , Bortezomib , Línea Celular Tumoral , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Morfolinas/farmacología , FN-kappa B/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño/metabolismo
8.
J Hepatol ; 50(3): 518-27, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19155085

RESUMEN

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and novel therapies are urgently needed. Recently, aberrant expression of Aurora kinases has been reported in various human cancers including HCC. We sought to investigate the potential of a potent and selective Aurora kinase inhibitor, VE-465, for targeted therapy of HCC. METHODS: Cytotoxicity effects of VE-465 were tested in Huh-7 and HepG2 cell lines. Inhibition of Aurora kinase activity was demonstrated by Western blotting and immunofluorescence staining. Mitotic perturbation was visualized by confocal microscopy. Cell cycle profiles and apoptosis were assessed by flow cytometry. In vivo efficacy was determined in nude mice with human HCC xenografts. RESULTS: We demonstrated that VE-465 induced proliferation blockade, histone H3 (Ser10) dephosphorylation, mitotic disturbance, endoreduplication, and apoptosis in Huh-7 and HepG2 cells. We also found that VE-465 suppressed Aurora kinase activity, prevented tumor growth, and induced apoptosis in a Huh-7 xenograft model. CONCLUSIONS: These findings show that VE-465 has potent anticancer effects in human HCC. Inhibitors of Aurora kinases may deserve further exploration as molecular targeted agents against HCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Piperazinas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Aurora Quinasas , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/patología , Mitosis/efectos de los fármacos
9.
Cancer Res ; 68(22): 9348-57, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010909

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death worldwide. Systemic treatments for HCC have been largely unsuccessful. OSU-03012 is a derivative of celecoxib with anticancer activity. The mechanism of action is presumably 3-phosphoinositide-dependent kinase 1 (PDK1) inhibition. This study investigated the potential of OSU-03012 as a treatment for HCC. OSU-03012 inhibited cell growth of Huh7, Hep3B, and HepG2 cells with IC(50) below 1 mumol/L. In Huh7 cells, OSU-03012 did not suppress PDK1 or AKT activity. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay and flow cytometry analysis indicated that OSU-03012 did not induce cellular apoptosis. Instead, morphologic studies by light and electron microscopy, as well as special biological staining with monodansylcadaverine, acridine orange, and microtubule-associated protein 1 light chain 3, revealed OSU-03012-induced autophagy of Huh7 cells. This OSU-03012-induced autophagy was inhibited by 3-methyladenine. Moreover, reactive oxygen species (ROS) accumulation was detected after OSU-03012 treatment. Blocking ROS accumulation with ROS scavengers inhibited autophagy formation, indicating that ROS accumulation and subsequent autophagy formation might be a major mechanism of action of OSU-03012. Daily oral treatment of BALB/c nude mice with OSU-03012 suppressed the growth of Huh7 tumor xenografts. Electron microscopic observation indicated that OSU-03012 induced autophagy in vivo. Together, our results show that OSU-03012 induces autophagic cell death but not apoptosis in HCC and that the autophagy-inducing activity is at least partially related to ROS accumulation.


Asunto(s)
Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Biochem Biophys Res Commun ; 376(2): 283-7, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18782567

RESUMEN

Lung adenocarcinoma Cl1-5 cells were selected from parental Cl1-0 cells based on their high metastatic potential. In a previous study, CRMP-1, an invasion suppressor gene, was shown to be suppressed in Cl1-5 cells. However, the regulation of CRMP-1 expression has not been explored. In this study, we showed nuclear factor-kappaB controls CRMP-1 expression. The electromobility shift assay showed that while Cl1-0 cells exhibited low NF-kappaB activity in response to TNF-alpha, an abundance of basal and TNF-alpha-induced NF-kappaB-DNA complex was detected in Cl1-5 cells. Supershift-coupled EMSA and Western blotting of nuclear proteins, however, revealed p50 protein, but not classic p65/p50 heterodimer in the complex. ChIP and EMSA demonstrated that p50 binds to a kappaB site residing between -1753 and -1743 of the CRMP-1 promoter region. Transfection of antisense p50 gene into Cl1-5 cells increased the CRMP-1 protein level and decreased the invasive activity of Cl1-5 cells.


Asunto(s)
Adenocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Neoplasias Pulmonares/patología , Subunidad p50 de NF-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Adenocarcinoma/genética , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Humanos , Neoplasias Pulmonares/genética , Invasividad Neoplásica , Regiones Promotoras Genéticas , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
11.
Cancer Res ; 68(16): 6698-707, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18701494

RESUMEN

Bortezomib, a proteasome inhibitor, has been clinically approved for the treatment of myeloma and lymphoma. Here, we report a differential effect of bortezomib on apoptosis in four hepatocellular carcinoma (HCC) cell lines and identify the major molecular event that determines sensitivity. Although bortezomib inhibited proteasome activity to a similar extent in all HCC cell lines, it showed differential effects on their viability: Huh-7 (IC(50) 196 nmol/L), Sk-Hep1 (IC(50) 180 nmol/L), Hep3B (IC(50) 112 nmol/L), and resistant PLC5 (IC(50) >1,000 nmol/L). Bortezomib caused cell cycle arrest at G(2)-M phase in all HCC cells tested whereas apoptotic induction was found only in sensitive cells but not in PLC5 cells. No significant bortezomib-induced NF-kappaB changes were noted in Huh-7 and PLC5. Bortezomib down-regulated phospho-Akt (P-Akt) in a dose- and time-dependent manner in all sensitive HCC cells whereas no alterations of P-Akt were found in PLC5. Down-regulation of Akt1 by small interference RNA overcame the apoptotic resistance to bortezomib in PLC5 cells, but a constitutively activated Akt1 protected Huh-7 cells from bortezomib-induced apoptosis. Furthermore, bortezomib showed suppression of tumor growth with down-regulation of P-Akt in Huh-7 tumors but not in PLC5 tumors. Down-regulation of P-Akt represents a major molecular event of bortezomib-induced apoptosis in HCC cell lines and may be a biomarker for predicting clinical response to HCC treatment. Targeting Akt signaling overcomes drug resistance to bortezomib in HCC cells, which provides a new approach for the combinational therapy of HCC.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazinas/farmacología , Animales , Western Blotting , Bortezomib , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Regulación hacia Abajo , Citometría de Flujo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Inhibidores de Proteasoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 112(7): 2927-34, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18628489

RESUMEN

We have recently demonstrated that nuclear expression of BCL10 predicts Helicobacter pylori (HP) independence of early-stage gastric diffuse large B-cell lymphoma (DLBCL) with histologic evidence of mucosa-associated lymphoid tissue (MALT). In this study, we examined the role of B cell-activating factor of TNF family (BAFF) in mediating BCL10 nuclear translocation and HP independence of gastric DLBCL (MALT). We used immunohistochemistry and immunoblotting to measure the expression of BAFF, pAKT, BCL3, BCL10, and NF-kappaB. Transactivity of NF-kappaB was measured by electromobility shift assay. In lymphoma samples from 26 patients with gastric DLBCL (MALT), we detected aberrant expression of BAFF in 7 of 10 (70%) HP-independent and in 3 of 16 (18.8%) HP-dependent cases (P = .015). BAFF overexpression was associated with pAKT expression (P = .032), and nuclear expression of BCL3 (P = .014), BCL10 (P = .015), and NF-kappaB (P = .004). In B-cell lymphoma Pfeiffer cells, BAFF activated NF-kappaB and AKT; the activated NF-kappaB up-regulated BCL10, and the activated AKT caused formation of BCL10/BCL3 complexes that translocated to the nucleus. Inhibition of AKT by LY294002 (a PI3K inhibitor) blocked BCL10 nuclear translocation, NF-kappaB transactivity, and BAFF expression. Our results indicate that autocrine BAFF signal transduction pathways may contribute to HP-independent growth of gastric DLBCL (MALT).


Asunto(s)
Factor Activador de Células B/metabolismo , Helicobacter pylori/fisiología , Linfoma de Células B de la Zona Marginal/microbiología , Linfoma de Células B de la Zona Marginal/patología , Linfoma de Células B Grandes Difuso/microbiología , Linfoma de Células B Grandes Difuso/patología , Neoplasias Gástricas/microbiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Proteína 10 de la LLC-Linfoma de Células B , Proteínas del Linfoma 3 de Células B , Línea Celular Tumoral , Núcleo Celular/metabolismo , Activación Enzimática , Femenino , Humanos , Linfoma de Células B de la Zona Marginal/enzimología , Linfoma de Células B Grandes Difuso/enzimología , Masculino , Persona de Mediana Edad , Modelos Biológicos , FN-kappa B/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/patología , Factores de Transcripción/metabolismo , Regulación hacia Arriba
13.
Ann Surg ; 247(2): 265-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18216531

RESUMEN

BACKGROUND: Gastric mucosa-associated lymphoid tissue (MALT) lymphoma is characterized by multifocality of the tumors, which often scatter over the mucosa of the stomach and adjacent upper gastrointestinal tract, and is therefore theoretically not curable by surgical resection. METHODS: We conducted a long-term follow-up study of 14 patients who received surgical treatment for gastric MALT lymphoma. Tissues from the surgical margins of the resected stomach were analyzed by polymerase chain reaction-based amplification of the complementarity-determining region 3 of the IgH gene for the presence of residual tumors. T (11;18)(q21;q21), a marker of Helicobacter pylori-independent MALT lymphoma, was analyzed by reverse transcription polymerase chain reaction. All living patients were restaged, and rebiopsied if suspicious lesions were identified. RESULTS: At a median follow-up of 11.5 years, only 1 patient had evidence of tumor recurrence. Three patients with molecularly proven residual tumors in the surgical margin remained disease-free at 9.6 to 11.6 years. Five patients with t(11;18)(q21;q21) in the tumor cells were disease-free at 9.2 to 12.6 years. CONCLUSION: Our results indicate that surgical resection is a highly curative treatment for gastric MALT lymphoma, even for patients with residual tumor cells in the surgical margins, and for patients with H. pylori-independent tumors. A revisit of surgical treatment for gastric MALT lymphoma is mandatory.


Asunto(s)
Gastrectomía/métodos , Linfoma de Células B de la Zona Marginal/cirugía , Neoplasias Gástricas/cirugía , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Biopsia , Regiones Determinantes de Complementariedad/genética , Endoscopía Gastrointestinal , Femenino , Estudios de Seguimiento , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Linfoma de Células B de la Zona Marginal/diagnóstico , Linfoma de Células B de la Zona Marginal/genética , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Pronóstico , ARN Neoplásico/genética , Reoperación , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Factores de Tiempo
14.
Lung Cancer ; 53(3): 303-10, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16806572

RESUMEN

The current study aimed to immunohistochemically examine the tumor glucocorticoids receptor (GR) expression in patients with advanced non-small cell lung cancer (NSCLC) and to examine the effect of glucocorticoids (GCs) on the in vitro NSCLC cells growth and chemosensitivity. High GR expression was detected in 51% of the tumor specimens. The difference in tumor GR expression was not associated with cell type, gender, age, or stage. The outcome was significantly superior for patients whose tumor showed high GR expression compared to those with either low expression or non-expression. The median progression-free survival was 8.0 versus 5.6 months (p=0.039) and overall survival was 18.1 versus 10.2 months, (p=0.003), respectively. Almost all these patients have received GC as antiemetics or allergic preventive treatment during chemotherapy courses, therefore, the effect of GC on the chemosensitivity in vivo was not evaluable. However, in vitro cytotoxicity assay showed that dexamethasone (DEX) had heterogeneous effects on the growth and chemosensitivity of the NSCLC cell lines. These findings suggest that tumor samples express high levels of GR in about half of the patients with advanced NSCLC, and this high expression of GR may be associated with better outcome. The effect of GC treatment on the chemosensitivity in NSCLC patients remains to be established.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucocorticoides/farmacología , Neoplasias Pulmonares/metabolismo , Receptores de Glucocorticoides/biosíntesis , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proliferación Celular , Dexametasona/farmacología , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Inmunohistoquímica , Ligandos , Neoplasias Pulmonares/tratamiento farmacológico
15.
J Endocrinol ; 188(2): 311-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16461557

RESUMEN

Glucocorticoids (GCs) are commonly co-administered with cisplatin in the treatment of patients with carcinomas to prevent drug-induced allergic reaction, nausea and vomiting. Although GC receptor (GR) is ubiquitous in carcinoma cells and has been linked to signal transduction pathways pertinent to cell growth and apoptosis, little is known regarding the possible effect of GC on the chemosensitivity of carcinomas. Our previous study demonstrated that dexamethasone (DEX) enhances the cytotoxicity to cisplatin in a GR-rich human cervical carcinoma cell line, SiHa. In this study, we found that this cisplatin cytotoxicity-enhancing effect of DEX correlated well with its effect on abrogating the cisplatin-induced activation of nuclear factor kappa B (NF-kappaB). RU486, a structural homologue of DEX, partially reversed this cytotoxicity-enhancing effect of DEX, a finding consistent with the well-known partial reversing effect of RU486 on DEX-induced NF-kappaB suppression. Furthermore, expression of a dominant-negative truncated IkappaBalpha gene in SiHa cells completely abolished the cisplatin cytotoxicity-enhancing effect of DEX. Our data suggest that the specific action of DEX on GR may enhance the cytotoxicity of cisplatin in selected GR-rich cancer cells by suppressing NF-kappaB activation.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Cisplatino/farmacología , Dexametasona/farmacología , FN-kappa B/antagonistas & inhibidores , Proteínas de Ciclo Celular/análisis , Línea Celular Tumoral , Fosfatasa 1 de Especificidad Dual , Regulación de la Expresión Génica/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Humanos , Proteínas Inmediatas-Precoces/análisis , Mifepristona/farmacología , Proteínas Quinasas Activadas por Mitógenos/análisis , FN-kappa B/metabolismo , Fosfoproteínas Fosfatasas/análisis , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/análisis , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Receptores de Glucocorticoides/metabolismo , Proteína X Asociada a bcl-2/análisis
16.
J Biol Chem ; 281(1): 167-75, 2006 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-16280327

RESUMEN

Bcl10 overexpression and nuclear translocation were originally identified in mucosa-associated lymphoid tissue lymphoma with t(1;14)(p32;q32) chromosome translocation. DNA amplification of Bcl10 was also found in other solid tumors. We have recently shown that nuclear translocation of Bcl10 is a specific molecular determinant of Helicobacter pylori-independent mucosa-associated lymphoid tissue lymphoma (Kuo, S.-H., Chen, L. T., Yeh, K.-H., Wu, M. S., Hsu, H. C., Yeh, P. Y., Mao, T. L., Chen, C. L., Doong, S. L., Lin, J. T., and Cheng, A.-L. (2004) J. Clin. Oncol. 22, 3491-3497). However, the molecular mechanism of Bcl10 nuclear translocation remains unknown. In this study, we observed that tumor necrosis factor-alpha (TNFalpha) up-regulates the expression of Bcl10 and induces a fraction of Bcl10 nuclear translocation in human breast carcinoma MCF7 cells. Chromatin immunoprecipitation assays and electrophoretic mobility shift assays indicated that an NF-kappaB-binding site resides in the Bcl10 5 '-untranslated region. This study also demonstrates that Akt1, activated by TNFalpha, phosphorylates Bcl10 at Ser218 and Ser231 and that phosphorylated Bcl10 subsequently complexes with Bcl3 to enter the nucleus. Either inhibition of Akt1 or depletion of Bcl3 blocks Bcl10 nuclear translocation. In summary, these findings characterize a molecular linkage that directs Bcl10 nuclear translocation in response to TNFalpha treatment.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Adaptadoras Transductoras de Señales/química , Proteína 10 de la LLC-Linfoma de Células B , Proteínas del Linfoma 3 de Células B , Línea Celular Tumoral , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , FN-kappa B/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo , Factores de Transcripción , Activación Transcripcional/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
17.
Oncology ; 68(4-6): 538-47, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16037687

RESUMEN

OBJECTIVE: Epidermoid growth factor receptor (EGFR, HER1) is overexpressed in a majority of head-and-neck cancers, including nasopharyngeal carcinoma (NPC). Although EGFR inhibitors appear to be effective for some head-and-neck cancers, their efficacy in NPC remains unclear. METHODS: The effect of EGFR-specific tyrosine kinase inhibitors, including PD153035 and ZD1839, were studied in NPC-TW01, NPC-TW04, and HONE1 cell lines. The effect of combining EGFR inhibitors with cytotoxic agents was evaluated in NPC-TW04 cells. RESULTS: All three NPC cell lines expressed EGFR. PD153035 and ZD1839 inhibited the growth of NPC cells with IC50s around 10 and 20 microM, respectively. These inhibitors, however, effectively suppressed ligand-stimulated EGFR activation in NPC cells with a much lower concentration (> or =0.1 microM). The growth-suppression activity of EGFR inhibitors was closely associated with suppression of AKT phosphorylation. LY294002, a phosphatidylinositol-3 kinase (P13K)/AKT inhibitor, did suppress the growth of NPC cells. Pretreatment of EGFR inhibitors by 24 h significantly enhanced the cytotoxic effect of doxorubicin, paclitaxel, cisplatin, and 5-fluororuacil in NPC-TW04 cells. CONCLUSIONS: Our data indicate that inhibition of EGFR activation is not sufficient to induce growth inhibition in NPC cells in vitro. EGFR inhibitors may be useful adjuncts in treating NPC when combined with conventional anticancer drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Neoplasias Nasofaríngeas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Cisplatino/administración & dosificación , Doxorrubicina/administración & dosificación , Quimioterapia Combinada , Receptores ErbB/genética , Receptores ErbB/metabolismo , Fluorouracilo/administración & dosificación , Humanos , Técnicas In Vitro , Neoplasias Nasofaríngeas/patología , Paclitaxel/administración & dosificación , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Células Tumorales Cultivadas
18.
World J Gastroenterol ; 11(40): 6373-80, 2005 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-16419168

RESUMEN

AIM: To determine how glucocorticoids (GCs) may affect the growth and chemosensitivity of common carcinoma cells. METHODS: The effect of dexamethasone (DEX) on growth and chemosensitivity was assessed in 14 carcinoma cell lines. The function of GC receptors (GR) was assessed by MMTV reporter assay. Overexpression of GR was done by in vitro transfection and expression of a GR-expressing vector. Immunohistochemical stain of tissues and cells were done by PA1-511A, an anti-GR monoclonal antibody. RESULTS: DEX inhibited cell growth of four (MCF-7, MCF-7/MXR1, MCF-7/TPT300, and HeLa), increased cisplatin cytotoxicity of one (SiHa), and decreased cisplatin cytotoxicity of two (H460 and Hep3B) cell lines. The GR content of the seven cell lines affected by DEX was significantly higher than those of the seven cell lines unaffected by DEX (5.2+/-2.5 x 10(4) sites/cell vs 1.3+/-1.4 x 10(4) sites/cell, P = 0.005). Only two DEX-unresponsive cell lines (NPC-TW01 and NPC-TW04) contained high GR amounts in the range (1.9-8.1 x 10(4) sites/cell) of the seven DEX-responsive cell lines. The GR function of NPC-TW01 and NPC-TW04, however, was found to be impaired. The importance of high cellular amount of GR in mediating DEX susceptibility of the cells was further exemplified by GR dose-dependent drug resistance to cisplatin of AGS, a cell line with low GR content and was unaffected by DEX before transfection of GR-expressing vector. Immunohistochemical studies of human cancer tissues showed that 5 of the 45 (11.1%) breast cancer and 43 of the 85 (50.6%) non-small cell lung cancer had high GR contents at the ranges of the GC-responsive carcinoma cell lines. CONCLUSION: The growth and chemosensitivity of human carcinomas with high GR contents may be affected by GC. However, in light of the heterogeneous and even contradictive effects of GC on these cells, routine examination of GR contents of human carcinoma tissues may not be clinically useful until other markers that help predict the ultimate effect of GC on individual patients are identified.


Asunto(s)
Antiinflamatorios/farmacología , Línea Celular Tumoral/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/farmacología , Receptores de Glucocorticoides/metabolismo , Carcinoma , Genes Reporteros , Humanos , Ligandos
19.
Anticancer Res ; 24(5A): 3035-40, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15517913

RESUMEN

BACKGROUND: Reactivation of HBV replication is a clinically significant complication in HBV(+) patients receiving chemotherapy. We recently found that nearly half of the HBV reactivation in lymphoma patients occurred within 2 weeks of the first dose of chemotherapy. We hypothesized that mechanisms other than immunosuppression, such as direct stimulation of HBV replication by anticancer drugs, might be involved in this type of HBV reactivation. MATERIALS AND METHODS: 2.2.15 cells, which secrete HBV particles constitutively, were used in the experiments. Real-time quantitative polymerase chain reaction was used to quantitate HBV DNA, and microparticle enzyme immunoassay to measure HBV surface antigen (HBsAg). RESULTS: HBV DNA secretion in culture medium was dose- dependently increased by doxorubicin, one of the most commonly used anticancer drugs for lmphoma. One-hour exposure of cells to 1 microM doxorubicin induced a 15.4+/-5.9-fold and a 3.05+/-0.09-fold increase of HBV DNA and HBsAg on the 4th culture day, respectively. Lamivudine suppressed the doxorubicin-induced increase of HBV DNA. CONCLUSION: Our data suggest that cytotoxic agents may stimulate the replication of HBVand thereby contribute to the reactivation of HBV during systemic chemotherapy. Importantly, this adverse effect of cytotoxic agents may be preventable by co-administration of lamivudine.


Asunto(s)
Doxorrubicina/efectos adversos , Virus de la Hepatitis B/fisiología , Hepatoblastoma/tratamiento farmacológico , Hepatoblastoma/virología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/virología , Activación Viral/efectos de los fármacos , Antibióticos Antineoplásicos/efectos adversos , Línea Celular Tumoral , ADN Viral/metabolismo , Relación Dosis-Respuesta a Droga , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/complicaciones , Hepatitis B Crónica/virología , Humanos , Cinética , Lamivudine/farmacología , Reacción en Cadena de la Polimerasa
20.
J Clin Oncol ; 22(17): 3491-7, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15337797

RESUMEN

PURPOSE: A high percentage of early-stage, high-grade gastric mucosa-associated lymphoid tissue (MALT) lymphomas remain Helicobacter pylori dependent. t(11;18)(q21;q21), a genetic aberration highly predictive of H. pylori-independent status in low-grade gastric MALT lymphoma, is rarely detected in its high-grade counterpart. This study examined whether nuclear expression of BCL10 or nuclear factor kappa B (NF-kappaB) is useful in predicting H. pylori-independent status in patients with stage IE high-grade gastric MALT lymphomas. PATIENTS AND METHODS: Twenty-two patients who had participated in a prospective study of H. pylori eradication for stage IE high-grade gastric MALT lymphomas were studied. The expression of BCL10 and NF-kappaB in pretreatment paraffin-embedded lymphoma tissues was evaluated by immunohistochemistry and confocal immunofluorescence microscopy. The presence of t(11;18)(q21;q21) was identified by a multiplex reverse transcriptase polymerase chain reaction of the API2-MALT1 chimeric transcript. RESULTS: Aberrant nuclear expression of BCL10 was detected in seven (87.5%) of eight H. pylori-independent and in none of 14 H. pylori-dependent high-grade gastric MALT lymphomas (P <.001). All seven patients with nuclear BCL10 expression had nuclear expression of NF-kappaB, compared with only two of 15 patients without nuclear BCL10 expression (P =.002). As a single variable, the frequency of nuclear expression of NF-kappaB was also significantly higher in H. pylori-independent tumors than in H. pylori-dependent tumors (seven of eight [87.5%] v two of 15 [12.3%]; P =.002). The API2-MALT1 fusion transcript was detected in only one (12.5%) of eight H. pylori-independent lymphomas. CONCLUSION: Nuclear expression of BCL10 or NF-kappaB is highly predictive of H. pylori-independent status in high-grade gastric MALT lymphoma, and coexpression of these two markers in the nuclei is frequent.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Linfoma de Células B de la Zona Marginal/metabolismo , Linfoma no Hodgkin/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Adulto , Anciano , Proteína 10 de la LLC-Linfoma de Células B , Biomarcadores/análisis , Femenino , Helicobacter pylori , Humanos , Inmunohistoquímica , Linfoma de Células B de la Zona Marginal/virología , Linfoma no Hodgkin/virología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...