Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Carcinog ; 53 Suppl 1: E72-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23625650

RESUMEN

The extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK) pathway is an important cell proliferation pathway. We previously reported that the transport protein particle complex 4 (TRAPPC4), ERK2 interaction may activate ERK1/2, modulate pERK2 nuclear localization and regulate proliferation and apoptosis in colorectal cancer (CRC) cells. The present study further investigated the function of the TRAPPC4-ERK2 interaction in CRC in vitro and in vivo. Silencing of TRAPPC4 induced G0/G1 phase cell cycle arrest, upregulated p21 and downregulated cyclin B1 in CRC cells. Overexpression of TRAPPC4 after ERK2 silencing decreased the percentage of G0/G1 phase cells, increased the percentage of G2/M and S phase cells, downregulated p21, upregulated cyclin B1, and enhanced CRC cell viability. Immunohistochemical staining revealed that knockdown of TRAPPC4 downregulated pERK2, whereas overexpression of TRAPPC4 upregulated pERK2. Epidermal growth factor (EGF) stimulated upregulation of TRAPPC4 and pERK2 in SW1116 cells; EGF stimulation or overexpression of TRAPPC4 induced pERK2 nuclear translocation. Silencing of TRAPPC4 reduced SW1116 xenograft tumor growth in vivo, whereas overexpression of TRAPPC4 increased tumor growth, compared to control tumors. Moreover, modulation of TRAPPC4 expression in vivo affected the levels of pERK2 in the cytoplasm and nucleus and expression of p21. These results conclusively demonstrate that TRAPPC4 regulates ERK2 activation and also affects the distribution of activated pERK2 in CRC cells. The ability of ERK2 to play a role in colorectal carcinogenesis depends, at least in part, on TRAPPC4.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteínas de Transporte Vesicular/fisiología , Animales , Apoptosis , Western Blotting , Ciclo Celular , Núcleo Celular/metabolismo , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Factor de Crecimiento Epidérmico/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas , Proteínas de Transporte Vesicular/antagonistas & inhibidores
2.
Carcinogenesis ; 32(8): 1207-15, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21665895

RESUMEN

Aberrant methylation has been shown to trigger the inactivation of tumor suppressor genes during tumorigenesis. MicroRNAs (miRNAs) have been found deregulated in human colorectal cancer (CRC), and some of them may function as tumor suppressor genes. Here, we investigated CpG island promoter hypermethylation as a potential mechanism underlying miRNA disruption and identifed methylation-sensitive miRNAs that might repress CRC development. We compared differential expression of miRNAs after 5-aza-2'-deoxycitidine (5-aza-dC) treatment using microarrays. DNA methylation status of the candidate miRNA was analyzed. The candidate miRNA was transfected into CRC cells and growth-suppressive mechanisms were explored. Luciferase reporter assay and western blot were used to identify the target genes of the candidate miRNA. The expression of mir-345 was significantly increased after 5-aza-dC treatment. DNA methylation analyses of mir-345 showed high methylation levels in tumor versus normal tissues. Expression of mir-345 was significantly down-regulated in 51.6% of CRC tissues compared with corresponding non-cancerous tissues. Low expression of mir-345 was associated with lymph node metastasis and worse histological type. Increased mir-345 function was sufficient to suppress colon cancer cell proliferation and invasiveness in vitro. Furthermore, we identified BCL2-associated athanogene 3 (BAG3), an anti-apoptosis protein, to be a target of mir-345. These results suggested as a methylation-sensitive miRNA in CRC, mir-345 may play an important role of antineoplastic as a growth inhibitor in the development of CRC.


Asunto(s)
Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , MicroARNs/fisiología , Adenocarcinoma/genética , Adenocarcinoma/patología , Adulto , Anciano , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Movimiento Celular , Inmunoprecipitación de Cromatina , Islas de CpG , Cartilla de ADN , Decitabina , Femenino , Perfilación de la Expresión Génica , Genes bcl-2 , Humanos , Técnicas para Inmunoenzimas , Luciferasas/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
3.
Cancer Invest ; 27(3): 273-85, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19194827

RESUMEN

The role of the mTOR signal pathway in colorectal cancer (CRC) pathogenesis remains unclear, and the combination effect of PD98059 (an inhibitor for MEK) and rapamycin (an inhibitor for mTOR) on CRC is still unknown. Here, we found that combination treatment with PD98059 and rapamycin suppressed the proliferation of CRC cells, induced apoptosis, arrested cell cycle, and reduced the incidence and volume of CRC in mice, as well as inhibited phosphorylation of mTOR and the MEK signal pathway components, of which the effects were more significant than single-drug treatments. These findings indicate that PD98059 combined with rapamycin appears to be a promising strategy for inhibiting the initiation, and progression of CRC, which may provide a novel strategy for CRC prevention.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Flavonoides/administración & dosificación , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas/fisiología , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Serina-Treonina Quinasas TOR
4.
Int J Colorectal Dis ; 24(6): 629-39, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19229543

RESUMEN

BACKGROUND AND AIMS: Although the anticancer effects of rapamycin (RPM) and 5-aza-deoxycytidine (AZA) have been studied extensively, the combined effect of these two drugs on colorectal cancer (CRC) is still unknown. This study addresses the effect of AZA and RPM combination therapy on CRC and its influence on the mammalian target of rapamycin (mTOR) and its signal transduction pathway. SUBJECTS AND METHODS: Human CRC cell line HCT116 was treated with AZA alone, RPM alone, or concurrently with a combination of both drugs. Cell viability, apoptosis, and cell cycle distribution were analyzed. CRC was initiated in S-ICR mice, which were then treated with the drugs mentioned above, and tumor incidence and volume were measured. The activity of the mTOR signal transduction pathway was detected by Western blot analysis or immunohistochemistry. RESULTS: Combination treatment with AZA and RPM inhibited the growth of HCT116 cells, induced apoptosis, arrested the cell cycle, and reduced the incidence and tumor volume of CRC in mice, as well as inhibited the phosphorylation of components of the mTOR signal transduction pathway. These effects were more significant than those of single-drug treatments. CONCLUSION: Combination treatment with AZA and RPM inhibits the formation and growth of CRC. These findings may provide a novel strategy for CRC treatment.


Asunto(s)
Azacitidina/farmacología , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Células HCT116 , Humanos , Inmunohistoquímica , Ratones , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR , Carga Tumoral/efectos de los fármacos
5.
Cancer Invest ; 26(3): 317-25, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18317973

RESUMEN

To evaluate the effect of adjuvant chemotherapy on survival rates after curative resection of gastric cancer, MEDLINE (1969-2006), EMBASE (1974-2006), bibliographies, and review articles were searched for relevant articles. The meta-analysis was finally based on 15 trials that included 3212 patients. RR for death in the treated group was 0.90 (95% CI, 0.84-0.96) (P = 0.0010). Little or no significant benefits were suggested in subgroup analyses between different population and regimens either. Postoperative adjuvant chemotherapy for gastric cancer confers slightly significant benefits compared to the surgery only group. The prognoses after surgery may differ among various population groups or because of different regimens.


Asunto(s)
Quimioterapia Adyuvante , Neoplasias Gástricas/tratamiento farmacológico , Terapia Combinada , Procedimientos Quirúrgicos del Sistema Digestivo , Humanos , Periodo Posoperatorio , Ensayos Clínicos Controlados Aleatorios como Asunto , Neoplasias Gástricas/cirugía
6.
Zhonghua Zhong Liu Za Zhi ; 29(12): 889-93, 2007 Dec.
Artículo en Zh | MEDLINE | ID: mdl-18478925

RESUMEN

OBJECTIVE: To investigate the synergistic effect of rapamycin (RPM) and PD98059 on human colorectal cancer cells and its potential mechanisms. METHODS: Three human colorectal cancer cell lines SW480, HCT116 and HT29 were treated with RPM 10 nmol/L, PD98059 (10 micromol/L, 20 micromol/L, 40 micromol/L, 50 micromol/L), or RPM plus PD98059, respectively, and the sensitivity was analyzed by MTT assay. The cell cycle progression was evaluated by flow cytometry. Western blotting analysis was performed to examine the total and phosphorylated levels of mammalian target of rapamycin (mTOR) and its downstream translational signaling intermediates, 70 kDa ribosomal protein S6 kinase (p70s6k) and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1). RESULTS: Both RPM and PD98059 could inhibit viability of the three cell lines. The anti-proliferative effect of PD98059 exhibited a time/dose dependent manner and was strengthen by RPM. All the treatment with RPM, PD98059, and RPM + PD98059 induced arrest of cell cycle, although the arrest was confined at different cell cycle phases. In addition to their effect on proliferation and cell cycle, both inhibitors also reduced phosphorylation levels of mTOR, p70s6k, and 4E-BP1, as well as total 4E-BP1 levels in SW480 and HCT116 cells. That effect was reinforced when cells were treated with RPM plus PD98059 simultaneously, whereas total protein levels of mTOR and p70s6k remained unchanged. CONCLUSION: RPM and PD98059 inhibit proliferation of colorectal cancer cells synergistically, and induce cell cycle arrest. The modulation of mammalian target of rapamycin signaling pathway is involved in its potential mechanisms.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Flavonoides/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Sirolimus/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antibióticos Antineoplásicos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas de Ciclo Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Sinergismo Farmacológico , Células HCT116 , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR
7.
J Natl Cancer Inst ; 105(22): 1738-49, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24104608

RESUMEN

BACKGROUND: The molecular mechanisms that control the aggressiveness of gastric cancer (GC) remain poorly defined. Here we show that synbindin contributes to the aggressiveness of GC by activating extracellular signal-regulated protein kinase (ERK) signaling on the Golgi apparatus. METHODS: Expression of synbindin was examined in normal gastric mucosa (n = 44), intestinal metaplastic gastric mucosa (n = 66), and GC tissues (n=52), and the biological effects of synbindin on tumor growth and ERK signaling were detected in cultured cells, nude mice, and human tissue samples. The interaction between synbindin and mitogen-activated protein kinase kinase (MEK1)/ERK was determined by immunofluorescence and fluorescence resonance energy transfer assays. The transactivation of synbindin by nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) was detected using luciferase reporter assay and chromatin immunoprecipitation. RESULTS: High expression of synbindin was associated with larger tumor size (120.8 vs 44.8 cm(3); P = .01), advanced tumor node metastasis (TNM) stage (P = .003), and shorter patient survival (hazard ratio = 1.51; 95% confidence interval [CI] = 1.01 to 2.27; P = .046). Synbindin promotes cell proliferation and invasion by activating ERK2 on the Golgi apparatus, and synbindin is directly transactivated by NF-κB. Synbindin expression level was statistically significantly higher in human GCs with activated ERK2 than those with low ERK2 activity (intensity score of 11.5, 95% CI = 10.4 to 12.4 vs intensity score of 4.6, 95% CI 3.9 to 5.3; P < .001). Targeting synbindin in xenograft tumors decreased ERK2 phosphorylation and statistically significantly reduced tumor volume (451.2mm(3), 95% CI = 328.3 to 574.1 vs 726.1mm(3), 95% CI = 544.2 to 908.2; P = .01). CONCLUSIONS: Synbindin contributes to malignant phenotypes of GC by activating ERK on the Golgi, and synbindin is a potential biomarker and therapeutic target for GC.


Asunto(s)
Aparato de Golgi/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proteínas de Transporte Vesicular/metabolismo , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Técnica del Anticuerpo Fluorescente , Mucosa Gástrica/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Aparato de Golgi/enzimología , Xenoinjertos , Humanos , Estimación de Kaplan-Meier , Luciferasas , Sustancias Luminiscentes , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Proteínas del Tejido Nervioso/genética , Oportunidad Relativa , Fosforilación , Análisis por Matrices de Proteínas , Estudios Retrospectivos , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/mortalidad , Activación Transcripcional , Regulación hacia Arriba , Proteínas de Transporte Vesicular/genética
8.
PLoS One ; 6(8): e23262, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21826244

RESUMEN

The trafficking protein particle complex 4 (TRAPPC4) is implicated in vesicle-mediated transport, but its association with disease has rarely been reported. We explored its potential interaction with ERK2, part of the ERK1/2 complex in the Extracellular Signal-regulated Kinase/ Mitogen-activated Protein Kinase (ERK-MAPK) pathway, by a yeast two-hybrid screen and confirmed by co-immunoprecipitation (Co-IP) and glutathione S-transferase (GST) pull-down. Further investigation found that when TRAPPC4 was depleted, activated ERK1/2 specifically decreased in the nucleus, which was accompanied with cell growth suppression and apoptosis in colorectal cancer (CRC) cells. Overexpression of TRAPPC4 promoted cell viability and caused activated ERK1/2 to increase overall, but especially in the nucleus. TRAPPC4 was expressed more highly in the nucleus of CRC cells than in normal colonic epithelium or adenoma which corresponded with nuclear staining of pERK1/2. We demonstrate here that TRAPPC4 may regulate cell proliferation and apoptosis in CRC by interaction with ERK2 and subsequently phosphorylating ERK1/2 as well as modulating the subcellular location of pERK1/2 to activate the relevant signaling pathway.


Asunto(s)
Núcleo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Neoplasias Colorrectales/genética , Humanos , Inmunohistoquímica , Inmunoprecipitación , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Interferencia de ARN , Análisis de Matrices Tisulares , Técnicas del Sistema de Dos Híbridos , Proteínas de Transporte Vesicular/genética
9.
Epigenetics ; 3(6): 330-5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19098451

RESUMEN

Colorectal cancer is a leading cause of morbidity and mortality worldwide, and its incidence has been increasing in recent years. The role of epigenetic modifications, including DNA methylation and histone modifications, has only recently been investigated. In this study, the effects of epigenetic agents such as folic acid (FA) and sodium butyrate (NaBu) on the development of colorectal cancer induced by 1,2-dimethylhydrazine (DMH) using ICR mice was examined. Of the mice treated in a chemopreventive manner with epigenetic agents, FA and NaBu, 15-50% developed colorectal cancer at 24 weeks compared with a 95% incidence of colorectal cancer in DMH-treated control mice. Folate deficiency can alter cytosine methylation in DNA leading to inappropriate activation of the proto-oncogene c-myc. We detected lower levels of p21(WAF1) gene expression in colorectal cancer samples, as well as significantly lower levels of acetylated histone H3, compared with samples from corresponding normal colorectal mucosa. In contrast, administration of NaBu increased levels of p21(WAF1) mRNA and p21(WAF1) protein, and was associated with an accumulation of histone acetylation. In summary, our results show that FA and NaBu reduce the incidence of colorectal cancer induced by DMH-induced in ICR mice, and therefore we hypothesize that targeting epigenetic targets should be further investigated for the prevention of colorectal cancer in humans.


Asunto(s)
Butiratos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Ácido Fólico/farmacología , Acetilación/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Neoplasias Colorrectales/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Metilación de ADN/efectos de los fármacos , Dieta , Suplementos Dietéticos , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Femenino , Ácido Fólico/sangre , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Ratones , Ratones Endogámicos ICR , Oportunidad Relativa , Regiones Promotoras Genéticas/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA