Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Oxid Med Cell Longev ; 2021: 8807676, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003521

RESUMEN

Warburgia ugandensis Sprague (W. ugandensis), widely distributed in Africa, is a traditional medicinal plant used for the treatment of various diseases including cancer. We intended to evaluate the anticolorectal cancer (CRC) activities of the crude extract from W. ugandensis (WUD) and reveal the underlying molecular mechanisms of its action. We found that WUD inhibited the proliferation of HT-29 and HCT116 cells in a time- and dose-dependent manner and induced intracellular ROS generation. The inhibitory effect of WUD on the proliferation of HT-29 and HCT116 cells could be attenuated by NAC (a ROS scavenger) in a dose-dependent manner. WUD induced G0/G1 phase arrest, down-regulated the protein expression of Cyclin D1 via ROS accumulation in HT-29 cells. In search of the molecular mechanism involved in WUD-induced Cyclin D1 down-regulation, it was found that WUD can suppress PI3K/Akt/GSK3ß signaling pathway in HT-29 cells. Next, it was found that WUD also activated apoptosis, poly-ADP ribose polymerase 1 (PARP1) cleavage and down-regulated pro-caspase 3 in HT-29 and HCT116 cells. Besides, WUD decreased the growth of colon tumors in vivo in the xenograft mouse model. We demonstrated for the first time that ROS and their modulation in the corresponding intracellular signaling could play a significant role in the potential activity of WUD against CRC cells.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Neoplasias del Colon/genética , Fase G1/genética , Extractos Vegetales/química , Fase de Descanso del Ciclo Celular/genética , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/patología , Femenino , Células HT29 , Humanos , Ratones , Especies Reactivas de Oxígeno
2.
Iran Biomed J ; 24(5): 314-23, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32429645

RESUMEN

Background: The Peucedanum species have many pharmacological effects due to the presence of coumarins, flavonoids, phenolic compounds, and essential fatty acids in these species. In this study, for the first time, the anticancer activity of Peucedanum chenur methanolic extract via the induction of apoptosis and inhibition of invasion in HCT-116 human colon cancer cells was investigated. Methods: P. chenur methanolic extract effect on HCT-116 cells viability and antioxidant activity were evaluated using MTT assay, 1,1-Diphenyl-2-picrylhydrazyl, and iron chelating tests, respectively. Changes in mRNA expression level in a panel of relevant genes were assessed by the quantitative real-time PCR. Also, apoptosis was assessed by cell cycle analysis and Annexin V/PI (propidium iodide) method, and the effect on cell migration was tested using scratch test. Results: P. chenur methanolic extract increased significantly the expression of BAX while decreased the expression of BCL-2, AKT1, FAK, RhoA, and matrix metalloproteinase (MMP) genes compared to the control group. BAX/BCL-2 ratio and apoptosis elevated, whereas cell migration reduced significantly. Besides, our extract showed an appropriate antioxidant activity. Conclusion: P. chenur may be introduced as a new chemopreventive agent in medicine due to its notable power in terms of induction of apoptosis and inhibition of invasion.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Apiaceae/química , Apoptosis , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Componentes Aéreos de las Plantas/química , Antineoplásicos Fitogénicos/farmacología , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/metabolismo , Línea Celular Tumoral , Ensayos de Migración Celular , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , ADN de Neoplasias/metabolismo , Depuradores de Radicales Libres/farmacología , Fase G1/efectos de los fármacos , Fase G1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Quelantes del Hierro/farmacología , Metanol , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Picratos/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Extractos Vegetales/toxicidad
3.
Sci Rep ; 8(1): 9365, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29921924

RESUMEN

18ß-Glycyrrhetinic acid (GA) is the active ingredient of the traditional Chinese medicine, Glycyrrhrzae Radix et Rhizoma. Here, we explored the effects of GA on hepatocellular carcinoma (HCC) in vitro and in vivo and the underlying molecular mechanisms. We confirmed that GA suppressed proliferation of various HCC cell lines. Treatment of GA caused G0/G1 arrest, apoptosis and autophagy in HCC cells. GA-induced apoptosis and autophagy were mainly due to the unfolded protein response. We compared the roles of the ATF4/CHOP and IRE1α/XBP1s UPR pathways, which were both induced by GA. The ATF4/CHOP cascade induced autophagy and was indispensable for the induction of apoptosis in GA-treated HCC cells. In contrast, the IRE1α/XBP1s cascade protected HCC cells from apoptosis in vitro and in vivo induced by GA. Despite this, activation of autophagy protected HCC cells from apoptosis induced by GA. We concluded that pharmacological inhibition of autophagy or IRE1α may be of benefit to enhance the antitumor activity of GA.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Ácido Glicirretínico/análogos & derivados , Neoplasias Hepáticas/metabolismo , Respuesta de Proteína Desplegada/genética , Apoptosis/genética , Autofagia/genética , Carcinoma Hepatocelular/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Fase G1/efectos de los fármacos , Fase G1/genética , Ácido Glicirretínico/farmacología , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase de Descanso del Ciclo Celular/genética , Respuesta de Proteína Desplegada/efectos de los fármacos , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
4.
J Pharmacol Sci ; 134(3): 190-196, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28757172

RESUMEN

Blockade of EGFR with reversible EGFR tyrosine kinase inhibitors (TKIs) is considered the frontline strategy for advanced NSCLC with EGFR mutations. However, acquired resistance to EGFR-TKI has been observed, resulting in disease progression and limited clinical benefit. Polyphyllin VII is the main member of polyphyllin family, which has been demonstrated to show strong anticancer activity against carcinomas. The sensitizing effect and underlying mechanism of Polyphyllin VII against acquired EGFR-TKI resistant NSCLC are still unexplored. In the present study, we aim to examined the sensitizing effect of Polyphyllin VII to gefitinib by modulating P21 signaling pathway in gefitinib acquired resistant NSCLC in vitro and in vivo. Gefitinib sensitive PC-9 cells and gefitinib acquired resistant H1975 cells were used. Cell proliferation and Clonogenic assay, Cell cycle analysis, Western blotting analysis and xenograft treatment were carried out. Polyphyllin VII enhanced the anti-proliferative effects of gefitinib and gefitinib-induced G1 phase arrest by modulation of P21 signaling pathway in acquired gefitinib resistant cells in vitro and in vivo. Polyphyllin VII elevated sensitization of gefitinib acquired resistant NSCLC cells to gefitinib through G1 phase arrest and modulation of P21 signaling pathway. It provides a potential new strategy to overcome gefitinib acquired resistance for EGFR-TKI resistant NSCLC.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Saponinas/farmacología , Saponinas/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Fase G1/efectos de los fármacos , Fase G1/genética , Gefitinib , Humanos , Neoplasias Pulmonares/genética , Fitoterapia , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
5.
Nucleic Acids Res ; 45(5): 2472-2489, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-27924000

RESUMEN

The biogenesis of ribosomes in vivo is an essential process for cellular functions. Transcription of ribosomal RNA (rRNA) genes is the rate-limiting step in ribosome biogenesis controlled by environmental conditions. Here, we investigated the role of folate antagonist on changes of DNA double-strand breaks (DSBs) landscape in mouse embryonic stem cells. A significant DSB enhancement was detected in the genome of these cells and a large majority of these DSBs were found in rRNA genes. Furthermore, spontaneous DSBs in cells under folate deficiency conditions were located exclusively within the rRNA gene units, representing a H3K4me1 hallmark. Enrichment H3K4me1 at the hot spots of DSB regions enhanced the recruitment of upstream binding factor (UBF) to rRNA genes, resulting in the increment of rRNA genes transcription. Supplement of folate resulted in a restored UBF binding across DNA breakage sites of rRNA genes, and normal rRNA gene transcription. In samples from neural tube defects (NTDs) with low folate level, up-regulation of rRNA gene transcription was observed, along with aberrant UBF level. Our results present a new view by which alterations in folate levels affects DNA breakage through epigenetic control leading to the regulation of rRNA gene transcription during the early stage of development.


Asunto(s)
Roturas del ADN de Doble Cadena , Deficiencia de Ácido Fólico/genética , Regulación del Desarrollo de la Expresión Génica , Genes de ARNr , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Células Madre Embrionarias/metabolismo , Feto/metabolismo , Antagonistas del Ácido Fólico/toxicidad , Deficiencia de Ácido Fólico/metabolismo , Fase G1/genética , Histonas/metabolismo , Leucovorina/farmacología , Metotrexato/toxicidad , Ratones , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo
6.
J Transl Med ; 13: 56, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25740019

RESUMEN

BACKGROUND: Although metastasis of clear cell renal cell carcinoma (ccRCC) is predominantly observed in late stage tumors, early stage metastasis of ccRCC can also be found with indefinite molecular mechanism, leading to inappropriate clinical decisions and poor prognosis. Stanniocalcin-1 (STC1) is a glycoprotein hormone involved in calcium/phosphate homeostasis, which regulates various cellular processes in normal development and tumorigenesis. This study aimed to investigate the role and mechanism of regulation of STC1 in the metastasis of early stage ccRCC. METHODS: STC1 mRNA and protein expression was determined in ccRCC surgical specimens, RCC cell lines, and human kidney tubule epithelial cell line HKC by real-time polymerase chain reaction (RT-PCR) and western blotting. Immunohistochemistry staining (IHC) and immunofluorescence were also used to examine the expression and localization of STC1 in ccRCC tissues and cancer cells. Knockdown and overexpression studies were conducted in vitro in RCC cell lines using small interfering RNAs (siRNA) and lentiviral-mediated gene delivery to evaluate the role of STC1 in cell proliferation, anchorage-dependent and independent growth, cell cycle control, and migration and invasion. RESULTS: STC1 mRNA and protein expression were significantly up-regulated in tumors when compared with non-tumor tissues, with the greatest increase in expression observed in metastatic tissues. Clinicopathological analysis revealed that STC1 mRNA expression was associated with Fuhrman tumor grade (P = 0.008) and overall Tumor Node Metastasis (TNM) staging (P = 0.018). STC1 expression was elevated in T1 stage metastatic tumors when compared with localized tumors, and was positively correlated with average tumor diameter. Silencing of STC1 expression by Caki-1 and A498 resulted in the inhibition of cell proliferation, migration, and invasion, meanwhile down-regulation of STC1 impaired epithelial-mesenchymal transition (EMT) of ccRCC cell lines. Overexpression of STC1 in Caki-2 enhanced cell growth and proliferation but not migration and invasion. Further investigation identified hypoxia and HIF-1α as candidate regulators of STC1 expression. CONCLUSIONS: Our findings demonstrate a role for STC1 in metastasis of early stage ccRCC and suggest that STC1 may be a biomarker of potential value both for the prognosis of this disease and for guiding clinical decisions regarding surgical strategies and adjuvant treatment.


Asunto(s)
Carcinoma de Células Renales/patología , Glicoproteínas/metabolismo , Neoplasias Renales/patología , Carcinoma de Células Renales/genética , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Fase G1/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glicoproteínas/genética , Humanos , Neoplasias Renales/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase S/genética
7.
Biomed Pharmacother ; 67(6): 489-96, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23773852

RESUMEN

Recent studies have demonstrated that natural water that has 65% of the deuterium concentration depleted, can exhibit anti-tumor properties. However, the anti-tumor effects of DDW on various nasopharyngeal carcinoma (NPC) cells have not previously been reported. In the present study, NPC cell lines and normal preosteoblast MC3T3-E1 cells were grown in RPMI1640 media containing different deuterium concentrations (50-150 ppm). The effects of DDW on the proliferation and migration of NPC and MC3T3-E1 cells were investigated using the MTT, plate colony formation, and Transwell assays, as well as Boyden chamber arrays, flow cytometry (FCM), western blot and immunofluorescence. We found that DDW was an effective inhibitor of NPC cell proliferation, plated colony formation, migration and invasion. In contrast, the growth of normal preosteoblast MC3T3-E1 cells was promoted when they were cultured in the presence of DDW. Cell cycle analysis revealed that DDW caused cell cycle arrest in the G1/S transition, reduced the number of cells in the S phase and significantly increased the population of cells in the G1 phase in NPC cells. Western blot analysis revealed that treatment with DDW significantly increased the expression of NADPH:quinone oxidoreductase-1 (NQO1), while immunofluorescence assay analysis revealed that treatment with DDW decreased the expression of PCNA and matrix metalloproteinase 9 (MMP9) in NPC cells. These results demonstrated that DDW is a novel, non-toxic adjuvant therapeutic agent that suppresses NPC cell proliferation, migration, and invasion by inducing the expression of NQO1 and causing cell cycle arrest, as well as decreasing PCNA and MMP9 expression.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Deuterio/metabolismo , Deuterio/farmacología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Agua/farmacología , Animales , Carcinoma , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Fase G1/efectos de los fármacos , Fase G1/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , NADP/genética , NADP/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Fase S/efectos de los fármacos , Fase S/genética , Agua/metabolismo
8.
Mol Cell Biochem ; 382(1-2): 173-83, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23807740

RESUMEN

Gonolobus condurango plant extract is used as an anticancer drug in some traditional systems of medicine including homeopathy, but it apparently lacks any scientific validation. Further, no detailed study is available to suggest whether condurango-glycoside-A (CGA), a major ingredient of condurango serves as a potent anticancer compound. Therefore, we investigated apoptosis-inducing ability of CGA against cervix carcinoma cells (HeLa). ß-galactosidase-activity and DNA damage were critically studied at different time points; while induced DNA-damage was observed at 9-12th hours, senescence of cells appeared at a later stage (18th hour after CGA treatment), implicating thereby a possible role of DNA damage in inducing pre-mature cell senescence. Concurrently, the number of cells undergoing apoptosis increased along with increase in reactive oxygen species (ROS) generation. Expression of p53 was also up-regulated, indicating that apoptosis could have been mediated through p53 pathway. DCHFDA (4',6-Diamidino-2-phenylindole dihydrochloride) assay, acridine orange/ethidium bromide staining and annexin V/PI assay results collectively confirmed that apoptosis was induced by increased ROS generation. Reduction in proliferation of cells was further evidenced by the cell cycle arrest at G0/G1 stage. Expression profiles of certain relevant genes and proteins like p53, Akt, Bcl-2, Bax, cytochrome c and caspase 3 also provided evidence of ROS mediated p53 up-regulation and further boost in Bax expression and followed by cytochrome c release and activation of caspase 3. Overall results suggest that CGA initiates ROS generation, promoting up-regulation of p53 expression, thus resulting in apoptosis and pre-mature senescence associated with DNA damage.


Asunto(s)
Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Daño del ADN , Marsdenia/química , Pregnanos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Senescencia Celular/genética , Fase G1/efectos de los fármacos , Fase G1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Espectrometría de Masas , Pregnanos/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Fase de Descanso del Ciclo Celular/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
Carcinogenesis ; 34(8): 1881-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23615403

RESUMEN

Epidemiological and experimental studies suggest that fiber and phenolic compounds might have a protective effect on the development of colon cancer in humans. Accordingly, we assessed the chemopreventive efficacy and associated mechanisms of action of a lyophilized red grape pomace containing proanthocyanidin (PA)-rich dietary fiber [grape antioxidant dietary fiber (GADF)] on spontaneous intestinal tumorigenesis in the Apc(Min/+) mouse model. Mice were fed a standard diet (control group) or a 1% (w/w) GADF-supplemented diet (GADF group) for 6 weeks. GADF supplementation greatly reduced intestinal tumorigenesis, significantly decreasing the total number of polyps by 76%. Moreover, size distribution analysis showed a considerable reduction in all polyp size categories [diameter <1mm (65%), 1-2mm (67%) and >2mm (87%)]. In terms of polyp formation in the proximal, middle and distal portions of the small intestine, a decrease of 76, 81 and 73% was observed, respectively. Putative molecular mechanisms underlying the inhibition of intestinal tumorigenesis were investigated by comparison of microarray expression profiles of GADF-treated and non-treated mice. We observed that the effects of GADF are mainly associated with the induction of a G1 cell cycle arrest and the downregulation of genes related to the immune response and inflammation. Our findings show for the first time the efficacy and associated mechanisms of action of GADF against intestinal tumorigenesis in Apc(Min/+) mice, suggesting its potential for the prevention of colorectal cancer.


Asunto(s)
Antioxidantes/farmacología , Ciclo Celular/efectos de los fármacos , Fibras de la Dieta/farmacología , Poliposis Intestinal/tratamiento farmacológico , Poliposis Intestinal/inmunología , Vitis/química , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Peso Corporal/inmunología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/prevención & control , Suplementos Dietéticos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Fase G1/efectos de los fármacos , Fase G1/genética , Fase G1/inmunología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Poliposis Intestinal/genética , Poliposis Intestinal/metabolismo , Pólipos Intestinales/tratamiento farmacológico , Pólipos Intestinales/genética , Pólipos Intestinales/inmunología , Pólipos Intestinales/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Masculino , Ratones , Transcriptoma/efectos de los fármacos , Transcriptoma/inmunología
10.
Int J Mol Med ; 31(6): 1443-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23563707

RESUMEN

The aim of the present study was to investigate the effects of electroacupuncture (EA) on the proliferation of chondrocytes and the molecular mechanism(s) involved. Passage 2 chondrocytes were randomly divided into four groups and treated with EA or nocodazole. After treatment, cell proliferation was determined using an MTT assay and DNA staining followed by FACS. The mRNA expression levels of cyclin D1, cyclin-dependent kinase (CDK)4, CDK6, phosphorylated retinoblastoma (pRb) and P16 were detected by RT-PCR, and the protein levels of cyclin D1, CDK4, CDK6, pRb and P16 were detected by western blotting. EA treatment significantly increased cell viability in a time-dependent manner and decreased the number of G0/G1 and G2/M phase chondrocytes and increased the number of S phase cells. The mRNA and protein levels of cyclin D1, CDK4, CDK6, (p)Rb and P16 consistently demonstrated a reverse trend with the levels in the chondrocytes treated with nocodazole. The expression levels of cyclin D1, CDK4, CDK6 and Rb were higher in chondrocytes receiving EA treatment when compared to levels in the untreated cells while expression of P16 was lower. In conclusion, EA treatment promotes chondrocyte proliferation via promotion of G1/S checkpoint transition in the cell cycle dependent on the activity of the P16-cyclin D1-CDK4/6-pRb pathway and this may, in part, explain its clinical effect in the treatment of osteoarthritis.


Asunto(s)
Condrocitos/metabolismo , Electroacupuntura , Fase G1 , Fase S , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Citometría de Flujo , Fase G1/genética , Regulación de la Expresión Génica , Nocodazol/farmacología , ARN Mensajero/genética , Ratas , Fase S/genética , Factores de Tiempo
11.
Biochem Biophys Res Commun ; 431(1): 58-64, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23291187

RESUMEN

Poly(ADP-ribose) polymerase-2 (PARP-2) catalyzes poly(ADP-ribosyl)ation (PARylation) and regulates numerous nuclear processes, including transcription. Depletion of PARP-2 alters the activity of transcription factors and global gene expression. However, the molecular action of how PARP-2 controls the transcription of target promoters remains unclear. Here we report that PARP-2 possesses transcriptional repression activity independently of its enzymatic activity. PARP-2 interacts and recruits histone deacetylases HDAC5 and HDAC7, and histone methyltransferase G9a to the promoters of cell cycle-related genes, generating repressive chromatin signatures. Our findings propose a novel mechanism of PARP-2 in transcriptional regulation involving specific protein-protein interactions and highlight the importance of PARP-2 in the regulation of cell cycle progression.


Asunto(s)
Ciclo Celular/genética , Regulación de la Expresión Génica , Histonas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Fase G1/genética , Células HEK293 , Antígenos de Histocompatibilidad/metabolismo , Histona Desacetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Poli(ADP-Ribosa) Polimerasas/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Factor de Transcripción YY1/metabolismo
12.
Asian Pac J Cancer Prev ; 14(11): 6363-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24377533

RESUMEN

Atractylis lancea (Thunb.) DC. (AL), an important medicinal herb in Asia, has been shown to have anti-tumor effects on cancer cells, but the involved mechanisms are poorly understood. This study focused on potential effects and molecular mechanisms of AL on the proliferation of the Hep-G2 liver cancer cell line in vitro. Cell viability was assessed by MTT test in Hep-G2 cells incubated with an ethanol extract of AL. Then, the effects of AL on apoptosis and cell cycle progression were determined by flow cytometry. Telomeric repeat amplification protocol (TRAP) assays was performed to investigate telomerase activity. The mRNA and protein expression of human telomerase reverse transcriptase (hTERT) and c-myc were determined by real-time RT-PCR and Western blotting. Our results show that AL effectively inhibits proliferation in Hep-G2 cells in a concentration- and time-dependent manner. When Hep-G2 cells were treated with AL after 48h,the IC50 was about 72.1 µg/ mL. Apoptosis was induced by AL via arresting the cells in the G1 phase. Furthermore, AL effectively reduced telomerase activity through inhibition of mRNA and protein expression of hTERT and c-myc. Hence, these data demonstrate that AL exerts anti-proliferative effects in Hep-G2 cells via down-regulation of the c-myc/hTERT/ telomerase pathway.


Asunto(s)
Atractylis/química , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Telomerasa/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Fase G1/efectos de los fármacos , Fase G1/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Extractos Vegetales/química , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Telomerasa/metabolismo
13.
Invest New Drugs ; 30(5): 1820-9, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21870073

RESUMEN

Targeting androgen receptor (AR) signaling with agents distinct from current antagonist drugs remains a rational approach to the prevention and treatment of prostate cancer (PCa). Our previous studies have shown that decursin and isomer decursinol angelate (DA), isolated from the Korean medicinal herb Angelica gigas Nakai, interrupt AR signaling and possess anti-PCa activities in vitro. In the LNCaP PCa cell model, these pyranoccoumarin compounds exhibit properties distinct from currently used antagonists (e.g., Casodex). However, both are rapidly de-esterified to decursinol, a partial AR agonist. We report here that a synthetic decursin analog, decursinol phenylthiocarbamate (DPTC), has greater in vivo stability than the parent compounds. DPTC-decursinol conversion was undetectable in mice. Furthermore, in LNCaP cells, DPTC decreased prostate specific antigen (PSA) expression, down-regulated AR abundance and mRNA and inhibited AR nuclear translocation. The effect of DPTC on AR and PSA mRNA and protein abundance was also observed in VCaP cells expressing wild type AR. DPTC inhibited growth of both PCa cell lines through G(1) cell cycle arrest and apoptosis, as did decursin and DA. Furthermore, i.p. administration of DPTC for 3 weeks suppressed the expression of AR target genes probasin and Nkx3.1 in mouse prostate glands. Overall, our data suggest that DPTC represents a prototype lead compound for development of in vivo stable and active novel decursin analogs for the prevention or therapy of PCa.


Asunto(s)
Antagonistas de Receptores Androgénicos/síntesis química , Antagonistas de Receptores Androgénicos/farmacología , Benzopiranos/farmacología , Butiratos/farmacología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzopiranos/síntesis química , Benzopiranos/química , Butiratos/síntesis química , Butiratos/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Fase G1/efectos de los fármacos , Fase G1/genética , Humanos , Isotiocianatos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Fenilcarbamatos/síntesis química , Fenilcarbamatos/química , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Mensajero/genética , Distribución Aleatoria , Receptores Androgénicos/genética , Transducción de Señal/genética , Tiocarbamatos/síntesis química , Tiocarbamatos/química
14.
Int Immunopharmacol ; 12(2): 334-41, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22182776

RESUMEN

Wogonin, one of flavonoid compounds isolated from Chinese herbal plants Scutellaria baicalensis Georgi, has been recognized as a potent anti-cancer agent acting through control of growth, differentiation and apoptosis. However, the underlying molecular mechanism of its anti-cancer activity remains to be further elucidated. In this study, we investigated the potential role of wogonin in the induced-apoptosis of human breast cancer cells MCF-7. Wogonin was found to inhibit the proliferation of MCF-7 in a concentration and time-dependent manner, notably wogonin could induce G1 phase arrest of MCF-7 cells. Wogonin-induced apoptosis was accompanied by a significant decrease of the Bcl-2 and survivin and increase of Bax and p53. Wogonin also increased active apoptosis forms of caspases-3, -8, -9 significantly. Z-DEVD-fmk, a specific caspase-3 inhibitor, significantly inhibited wogonin-induced cell apoptosis. Wogonin also suppressed the phosphorylation of PI3K/Akt and induced phosphorylation of ERK. PD98059, a specific ERK inhibitor, significantly blocked wogonin-induced apoptosis. On the other hand, LY294002, a specific PI3K inhibitor, significantly increased wogonin-induced cell apoptosis. Further study indicated that LY294002 not only down-regulated the expression of survivin alone, but also enhanced the inhibition of survivin expression combined with wogonin. In conclusion, the pro-apoptotic effect of wogonin is mediated through the activation of ERK and the activation of caspases, and is correlated with the block of the PI3K/Akt/survivin signal pathways in MCF-7 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Flavanonas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antineoplásicos/farmacología , Apoptosis/genética , Neoplasias de la Mama/genética , Caspasas/genética , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Medicamentos Herbarios Chinos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Flavonoides/farmacología , Fase G1/efectos de los fármacos , Fase G1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Survivin , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
15.
Anticancer Drugs ; 22(10): 986-94, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21822123

RESUMEN

The G(2) checkpoint is an indispensable pathway for cancers lacking p53 function, for delaying cell cycle progression, and for completing DNA repair. Therefore, disruption of this pathway is expected to offer selective therapy for these highly prevalent cancers. The aim of this study was to identify an inhibitor of the G(2) checkpoint including the ataxia-telangiectasia-mutated and Rad3-related checkpoint kinase 1 pathway that selectively suppresses the growth of p53-deficient cells. To obtain molecules with a novel mechanism of action, we constructed a high-throughput screening system that detected abrogation of the G(2) checkpoint in X-irradiated HT-29 cells. The screening resulted in identification of a guanidine analog, CBP-93872 that dose dependently inhibited the G(2) checkpoint induced by DNA damage. Interestingly, CBP-93872 directly suppressed the growth of p53-mutated cancer cell lines with wild-type CDKN2A by eliciting G(1) arrest, but not CDKN2A-deleted and/or wild-type p53 lines. CBP-93872 decreased phospho-cdc2 Y15 by inhibiting phosphorylation of Chk1, but did not suppress phospho-Chk2 or the kinase activities of either Chk1 or Chk2 in cellular or cell-free assays. These results suggest that a checkpoint modulator through suppression of Chk1 phosphorylation provides synthetic lethality to p53-deficient cells.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Fase G2/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Propanolaminas/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína Quinasa CDC2 , Camptotecina/farmacología , Proliferación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina B/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Fase G1/efectos de los fármacos , Fase G1/genética , Células HT29/efectos de los fármacos , Células HT29/efectos de la radiación , Humanos , Mutación , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Reproducibilidad de los Resultados
16.
Cancer Biol Ther ; 10(4): 344-50, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20543580

RESUMEN

Vitamin D derivatives can induce differentiation of human acute myeloid leukemia (AML) cells. Here, we investigated if the G1 cell cycle block associated with monocytic differentiation is modulated by the p53 status of the cells treated with 1,25D, alone or with plant antioxidants carnosic acid (C) or silibinin (S), and a p38 MAPK inhibitor SB202190 (SB), a combination (D-C/S-SB) previously shown to enhance differentiation of AML p53null cells. D-C/S-SB enhanced differentiation of OCI-AML3 (p53wt) and as expected HL60 (p53 null) cells, but not of MOLM-13 (p53wt) cells. Conversely, MOLM-13 (p53wt) cells treated with 1,25D and/or D-C/S-SB, resembled HL60 (p53 null) cells in rapid G1 block, while OCI-AML3 (p53wt) cells showed a delayed G1 block when treated in a similar way, indicating that there is no relationship between the p53 status and G1 block. Western blot analysis revealed that 1,25D and D-C/S-SB increased the inhibitory phosphorylation levels MEK-1 (P-Thr286), but decreased the levels of activated ERK1/2 (Thr202/Tyr204;Thr185/Tyr187), again without any apparent relationship to the p53 status. Interestingly, the increased levels of p21(Waf1/Cip1) were insufficient to promote a G1 block in this system, as only cell lines with increased levels of p27(Kip1) and p35Nck5a, an activator of Cdk5, showed a rapid G1 block. Overall, our data show that the p53-p21 axis is unlikely to have a role in differentiation-associated G1 block in AML cells with wt p53, and that this block is achieved by several, possibly co-operating but redundant pathways, that include inhibition of MEK-1 by p35Nck5a-activated Cdk5.


Asunto(s)
Antioxidantes/farmacología , Calcitriol/farmacología , Fase G1/efectos de los fármacos , Leucemia Mieloide Aguda , Proteína p53 Supresora de Tumor/metabolismo , Abietanos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Sinergismo Farmacológico , Fase G1/genética , Células HL-60 , Humanos , Imidazoles/farmacología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Extractos Vegetales/farmacología , Piridinas/farmacología , Silibina , Silimarina/farmacología
17.
Cancer Sci ; 101(4): 948-54, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20148895

RESUMEN

Arsenic trioxide (ATO), an ancient traditional Chinese medicine, has been successfully used as a therapeutic agent for leukemia. Drug resistance and toxicity are major concerns with the treatment. MicroRNAs (miRNAs) are endogenous small non-coding RNA molecules that might modulate cellular sensitivity to anticancer drugs. miRNA-21 (miR-21) is one of the most prominent miRNAs involved in various aspects of human cancers. However, miR-21 has been rarely characterized in chronic myelogenous leukemia (CML). Here, we used a specific anti-miR-21 oligonucleotide (AMO-miR-21) to sensitize K562 cells to ATO by degradation of miR-21. The results showed that both AMO-miR-21 and ATO caused growth inhibition, apoptosis, and G1-phase arrest in K562 cells. Meanwhile, AMO-miR-21 significantly promoted ATO-mediated growth inhibition and apotosis without affecting the G1 phase. Apoptotic cells were confirmed morphologically with Giemsa's staining. Furthermore, dual-luciferase reporter vector, containing two tandem miR-21 binding sites from PDCD4 3'UTR, validated that PDCD4 was directly regulated by miR-21. Therefore, AMO-miR-21 sensitized leukemic K562 cells to ATO by inducing apoptosis partially due to its up-regulation of PDCD4 protein level. The combination of ATO and AMO-miR-21 present therapeutic potential for CML.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , MicroARNs/farmacología , Oligonucleótidos/farmacología , Óxidos/uso terapéutico , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Trióxido de Arsénico , Ciclo Celular/genética , Fase G1/genética , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba
18.
Cancer Res ; 69(16): 6565-72, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19638577

RESUMEN

D- and E-type cyclins mediate G(1)-S phase cell cycle progression through activation of specific cyclin-dependent kinases (cdk) that phosphorylate the retinoblastoma protein (pRb), thereby alleviating repression of E2F-DP transactivation of S-phase genes. Cyclin D1 is often overexpressed in a variety of cancers and is associated with tumorigenesis and metastasis. Loss of cyclin D can cause G(1) arrest in some cells, but in other cellular contexts, the downstream cyclin E protein can substitute for cyclin D and facilitate G(1)-S progression. The objective of this study was to determine if a flexible heteroarotinoid anticancer compound, SHetA2, regulates cell cycle proteins and cell cycle progression in ovarian cancer cells. SHetA2 induced cyclin D1 phosphorylation, ubiquitination, and proteasomal degradation, causing G(1) arrest in ovarian cancer cells despite continued cyclin E2 expression and independently of p53 and glycogen synthase kinase-3beta. Cyclin D1 loss inhibited pRb S780 phosphorylation by cyclin D1-cdk4/6 and released p21 from cyclin D1-cdk4/6-p21 protein complexes to form cyclin E2-cdk2-p21 complexes, which repressed phosphorylation of pRb S612 by cyclin E2-cdk2 and ultimately E2F-DP transcriptional activity. G(1) arrest was prevented by overexpression or preventing degradation of cyclin D1 but not by restoration of pRb S612 phosphorylation through p21 knockdown. In conclusion, we show that loss of cyclin D1 in ovarian cancer cells treated with SHetA2 is sufficient to induce G(1) cell cycle arrest and this strategy is not impeded by the presence of cyclin E2. Therefore, cyclin D1 is a sufficient therapeutic target in ovarian cancer cells.


Asunto(s)
Proliferación Celular , Ciclina D1/metabolismo , Ciclinas/genética , Fase G1 , Neoplasias Ováricas/patología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proliferación Celular/efectos de los fármacos , Cromanos/farmacología , Ciclina D1/genética , Ciclina D1/fisiología , Evaluación Preclínica de Medicamentos , Femenino , Fase G1/efectos de los fármacos , Fase G1/genética , Fase G1/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/fisiología , Tionas/farmacología , Transfección , Células Tumorales Cultivadas , Ubiquitinación/efectos de los fármacos
19.
Mutagenesis ; 21(6): 405-10, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17065160

RESUMEN

Previous reports showed the protective effect of the synthetic antioxidant butylated hydroxytoluene (BHT) against the chromosomal damage induced by bleomycin (BLM), cadmium chloride and potassium dichromate. To test the hypothesis that this effect was exerted by inhibition and/or scavenging of reactive oxygen species (ROS), the effect of BHT on the chromosomal damage induced by a high dose-rate gamma rays (HDR (192)Ir). Experiments were carried out by irradiating G(1) CHO cells with nominal doses of 1, 2 or 3 Gy. BHT (doses of 1.0, 2.5 or 5.0 microg/ml) was added to the culture immediately before or immediately after irradiation. Cells were then incubated in the presence of BHT for 13 h until harvesting and fixation. Results obtained showed that BHT did not decrease the chromosomal damage induced by radiation in any consistent fashion. On the contrary, in cells post-treated with 5.0 microg/ml of BHT the yield of chromosomal aberrations increased in several experimental points. These results with ionizing radiation suggest that the previous observed protective effects of BHT on the chromosomal damage induced by chemical genotoxicants may not be mediated solely through the scavenging or inactivating reactive oxidative species. The decrease of the yield of chromosomal damage induced by BLM could be due to the union of BHT with a metallic ion, in this case Fe (II), required for the activation of BLM. In the same way, the protective effect of BHT on the chromosomal damage induced by cadmium chloride and potassium dichromate could be due to the decrease of the effective dose of both salts in the cell through the chelation of the cations by BHT.


Asunto(s)
Antioxidantes/farmacología , Hidroxitolueno Butilado/farmacología , Aberraciones Cromosómicas/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Rayos gamma/efectos adversos , Sustancias Protectoras/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Células CHO/efectos de los fármacos , Células CHO/efectos de la radiación , Cloruro de Cadmio/toxicidad , Aberraciones Cromosómicas/efectos de la radiación , Colorantes/toxicidad , Cricetinae , Fase G1/efectos de los fármacos , Fase G1/genética , Dicromato de Potasio/toxicidad , Especies Reactivas de Oxígeno/metabolismo
20.
J Neurosurg ; 98(2): 378-84, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12593626

RESUMEN

OBJECT: Ionizing radiation is the gold-standard adjuvant treatment for glioblastoma multiforme (GBM), the most aggressive primary brain tumor. The mechanisms underlying neoplastic glial cell growth inhibition after administration of ionizing radiation, however, remain largely unknown. In this report, the authors characterize the response of GBM cells to ionizing radiation and elucidate factors that correlate with the radiosensitivity of these tumors. METHODS: Six human GBM cell lines were subjected to increasing doses of radiation. Each demonstrated a dose-dependent suppression of cell proliferation. In the most radiosensitive cell line, the authors demonstrated a transient increase in the expression of the cyclin-dependent kinase inhibitors (CDKIs) p21 and p27, which corresponded with a G1 cell-cycle arrest. In contrast, the most radioresistant cell line demonstrated a decrease in p21 and p27 expression levels, which correlated with a failure to arrest. Apoptosis did not occur in any cell line following irradiation. Instead, autophagic cell changes were observed following administration of radiation, regardless of the relative radiosensitivity of the cell line. CONCLUSIONS: These findings elucidate some of the molecular responses of GBMs to irradiation and suggest novel targets for future therapy.


Asunto(s)
Autofagia/genética , Autofagia/efectos de la radiación , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Proteínas de Ciclo Celular/efectos de la radiación , Ciclinas/efectos de la radiación , Inhibidores Enzimáticos/efectos de la radiación , Fase G1/genética , Fase G1/efectos de la radiación , Glioblastoma/patología , Glioblastoma/radioterapia , Radiación Ionizante , Proteínas Supresoras de Tumor/efectos de la radiación , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/análisis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Ciclinas/análisis , Inhibidores Enzimáticos/análisis , Glioblastoma/genética , Humanos , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Orgánulos/genética , Orgánulos/efectos de la radiación , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , Factores de Tiempo , Células Tumorales Cultivadas/patología , Células Tumorales Cultivadas/efectos de la radiación , Proteínas Supresoras de Tumor/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA