Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Oncotarget ; 7(51): 85208-85219, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27863441

RESUMEN

Although the mechanistic target of rapamycin (mTOR) inhibitor, everolimus, has improved the outcome of patients with renal cell carcinoma (RCC), improvement is temporary due to the development of drug resistance. Since many patients encountering resistance turn to alternative/complementary treatment options, an investigation was initiated to evaluate whether the natural compound, sulforaphane (SFN), influences growth and invasive activity of everolimus-resistant (RCCres) compared to everolimus-sensitive (RCCpar) RCC cell lines in vitro. RCC cells were exposed to different concentrations of SFN and cell growth, cell proliferation, apoptosis, cell cycle, cell cycle regulating proteins, the mTOR-akt signaling axis, adhesion to human vascular endothelium and immobilized collagen, chemotactic activity, and influence on surface integrin receptor expression were investigated. SFN caused a significant reduction in both RCCres and RCCpar cell growth and proliferation, which correlated with an elevation in G2/M- and S-phase cells. SFN induced a marked decrease in the cell cycle activating proteins cdk1 and cyclin B and siRNA knock-down of cdk1 and cyclin B resulted in significantly diminished RCC cell growth. SFN also modulated adhesion and chemotaxis, which was associated with reduced expression of the integrin subtypes α5, α6, and ß4. Distinct differences were seen in RCCres adhesion and chemotaxis (diminished by SFN) and RCCpar adhesion (enhanced by SFN) and chemotaxis (not influenced by SFN). Functional blocking of integrin subtypes demonstrated divergent action on RCC binding and invasion, depending on RCC cell sensitivity to everolimus. Therefore, SFN administration could hold potential for treating RCC patients with established resistance towards everolimus.


Asunto(s)
Antineoplásicos/uso terapéutico , Isotiocianatos/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Apoptosis , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Adhesión Celular , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Ciclina B/genética , Ciclina B/metabolismo , Resistencia a Antineoplásicos , Everolimus/uso terapéutico , Humanos , Integrina alfa5/metabolismo , ARN Interferente Pequeño/genética , Sulfóxidos , Serina-Treonina Quinasas TOR/metabolismo
2.
J Pharmacol Sci ; 127(3): 332-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25837931

RESUMEN

Pine needle oil from crude extract of pine needles has been used as an anti-cancer agent in Traditional Chinese Medicine. The α-pinene is a natural compound isolated from pine needle oil which has been shown anti-cancer activity. In previous study, we found that pine needle oil exhibited significant inhibitory effect on hepatoma carcinoma BEL-7402 cells. In this study, we investigate the inhibition of α-pinene on hepatoma carcinoma BEL-7402 cells in vitro and in vivo and further explore the mechanism. The results show that liver cancer cell growth was inhibited obviously with inhibitory rate of 79.3% in vitro and 69.1% in vivo, Chk1 and Chk2 levels were upregulated, CyclinB, CDC25 and CDK1 levels were downregulated.


Asunto(s)
Antineoplásicos Fitogénicos , Carcinoma Hepatocelular/patología , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Neoplasias Hepáticas/patología , Puntos de Control de la Fase M del Ciclo Celular/genética , Monoterpenos/farmacología , Pinus/química , Aceites de Plantas/química , Animales , Monoterpenos Bicíclicos , Proteína Quinasa CDC2 , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2/metabolismo , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Ratones Desnudos , Monoterpenos/aislamiento & purificación , Trasplante de Neoplasias , Fitoterapia , Proteínas Quinasas/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
3.
Molecules ; 19(11): 17663-81, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25365293

RESUMEN

Flavonoids, containing mainly kaempferol rhamnohexoside derivatives, were extracted from Gynostemma pentaphyllum (G. pentaphyllum) and their potential growth inhibition effects against H460 non-small cell lung cancer cells was explored and compared to that on A549 cells. The extracted flavonoids were found to exhibit antiproliferation effects against H460 cells (IC50 = 50.2 µg/mL), although the IC50 of H460 is 2.5-fold that of A549 cells (IC50 = 19.8 µg/mL). Further investigation revealed that H460 cells are more susceptible to kaempferol than A549, whereas A549 cell growth is better inhibited by kaempferol rhamnohexoside derivatives as compared with H460. In addition, flavonoids from G. pentaphyllum induced cell cycle arrest at both S and G2/M phases with concurrent modulated expression of the cellular proteins cyclin A, B, p53 and p21 in A549 cells, but not H460. On the contrary, apoptosis and concomitant alteration in balance of BCL-2 and BAX expression as well as activation of caspase-3 were equally affected between both cells by flavonoid treatment. These observations strongly suggest the growth inhibition discrepancy between H460 and A549 following flavonoid treatment can be attributed to the lack of cell cycle arrest in H460 cells and the differences between H460 and A549 cells may serve as contrasting models for further mechanistic investigations.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Flavonoides/farmacología , Gynostemma/química , Extractos Vegetales/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Ciclina B/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Flavonoides/química , Humanos , Quempferoles/farmacología , Extractos Vegetales/química , Proteína p53 Supresora de Tumor/metabolismo
4.
Bioelectromagnetics ; 35(5): 337-46, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24619849

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exhibits its potent antitumor activity via membrane receptors on cancer cells without deleterious side effects for normal tissue. However, as many other cancer types, breast cancer cells develop a resistance to TRAIL. In the present study, we reported that exposure to 3.0 mT static magnetic field (SMF) mediated the sensitization of breast cancer cells to TRAIL-induced apoptosis. This effect was significantly reduced by the forced expression of survivin, suggesting the sensitization was mediated at least in part through the inhibition of survivin expression. In addition, SMF alone or in combination with TRAIL induced a cell cycle arrest within the G2 /M phase, and the reduction in the survivin protein level was associated with the downregulated expression of Cdc2, a cyclin B-dependent kinase that is necessary for the entry into the M phase. Taken together, our results demonstrated that SMF promoted TRAIL-induced apoptosis by inhibiting the expression of Cdc2 and, subsequently, survivin. Of note, SMF did not sensitize untransformed human mammary epithelial cells to TRAIL-mediated apoptosis. Therefore, the combined treatment of SMF and TRAIL may offer an attractive strategy for safely treating resistant breast cancers.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Proteína Quinasa CDC2/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Campos Magnéticos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Neoplasias de la Mama/terapia , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina B/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Magnetoterapia , Survivin
5.
Cell Death Dis ; 5: e1087, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24577085

RESUMEN

Cucurbitacin E (CuE) is a natural compound previously shown to have anti-feedant, antioxidant and antitumor activities as well as a potent chemo-preventive action against cancer. The present study investigates its anti-proliferative property using MTT assay; CuE demonstrated cytotoxic activity against malignant glioma GBM 8401 cells and induced cell cycle G2/M arrest in these cells. CuE-treated cells accumulated in metaphase (CuE 2.5-10 µM) as determined using MPM-2 by flow cytometry. We attempted to characterize the molecular pathways responsible for cytotoxic effects of CuE in GBM 8401 cells. We studied the genome-wide gene expression profile on microarrays and molecular networks by using pathway analysis tools of bioinformatics. The CuE reduced the expression of 558 genes and elevated the levels of 1354 genes, suggesting an existence of the common pathways involved in induction of G2/M arrest. We identified the RB (GADD45ß and GADD45γ) and the p53 (GADD45α) signaling pathways as the common pathways, serving as key molecules that regulate cell cycle. Results indicate that CuE produced G2/M arrest as well as the upregulation of GADD45 γ and binding with CDC2. Both effects increased proportionally with the dose of CuE, suggesting that the CuE-induced mitosis delay is regulated by GADD45γ overexpression. Our findings suggest that, in addition to the known effects on cancer prevention, CuE may have antitumor activity in glioma therapy.


Asunto(s)
Antimitóticos/farmacología , Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/metabolismo , Cucumis melo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitosis/efectos de los fármacos , Neuroblastoma/metabolismo , Triterpenos/farmacología , Antimitóticos/aislamiento & purificación , Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteína Quinasa CDC2 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Biología Computacional , Cucumis melo/química , Ciclina B/genética , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Relación Dosis-Respuesta a Droga , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neuroblastoma/genética , Neuroblastoma/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fitoterapia , Tallos de la Planta , Plantas Medicinales , Unión Proteica , Transducción de Señal/efectos de los fármacos , Triterpenos/aislamiento & purificación , Regulación hacia Arriba
6.
J Nat Med ; 68(1): 53-62, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23604974

RESUMEN

Berberine is an alkaloid isolated from the Chinese herbal medicine Huanglian, and has long been used as an antibiotic. Its antineoplastic properties were subsequently discovered in vitro. The purpose of this study was to investigate the effects of berberine on the growth of human colorectal carcinoma cells in vitro and in vivo. The results showed that berberine inhibited human colorectal adenocarcinoma (LoVo) cell growth in a time- and dose-dependent manner. A WST-1 assay showed that the IC50 value after 72 h was 40.79 ± 4.11 µM. Cell cycle analysis of 40 µM berberine-treated LoVo cells by flow cytometry showed accumulation of cells in the G2/M phase. The inhibition of LoVo cell growth by berberine was associated with the suppression of cyclin B1, cdc2, and cdc25c proteins. Berberine at a dose of 50 mg kg(-1) day(-1) showed inhibitory rates of 45.3% in a human colorectal adenocarcinoma xenograft in nude mice. The combination of berberine and 5-fluorouracil (5-FU) had a higher inhibitory rate (59.8%) than the berberine group (36.4%, P = 0.01), but no significant difference was observed between the 5-FU group (43.0%, P = 0.06) and the combination group. These results support the possibility that berberine may be useful as an alternative therapy for colorectal carcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Fitogénicos/farmacología , Berberina/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteína Quinasa CDC2 , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ciclina B/metabolismo , Ciclina B1/metabolismo , Quinasas Ciclina-Dependientes , Relación Dosis-Respuesta a Droga , Fluorouracilo/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Células HT29 , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , Fosfatasas cdc25/metabolismo
7.
Cell Cycle ; 12(16): 2598-607, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23907120

RESUMEN

Microtubule-poisoning drugs, such as Paclitaxel (or Taxol, PTX), are powerful and commonly used anti-neoplastic agents for the treatment of several malignancies. PTX triggers cell death, mainly through a mitotic arrest following the activation of the spindle assembly checkpoint (SAC). Cells treated with PTX slowly slip from this mitotic block and die by mitotic catastrophe. However, cancer cells can acquire or are intrinsically resistant to this drug, posing one of the main obstacles for PTX clinical effectiveness. In order to override PTX resistance and increase its efficacy, we investigated both the enhancement of mitotic slippage and the block of mitotic exit. To test these opposing strategies, we used physiological hyperthermia (HT) to force exit from PTX-induced mitotic block and the anaphase-promoting complex/cyclosome (APC/C) inhibitor, proTAME, to block mitotic exit. We observed that application of HT on PTX-treated cells forced mitotic slippage, as shown by the rapid decline of cyclin B levels and by microscopy analysis. Similarly, HT induced mitotic exit in cells blocked in mitosis by other antimitotic drugs, such as Nocodazole and the Aurora A inhibitor MLN8054, indicating a common effect of HT on mitotic cells. On the other hand, proTAME prevented mitotic exit of PTX and MLN8054 arrested cells, prolonged mitosis, and induced apoptosis. In addition, we showed that proTAME prevented HT-mediated mitotic exit, indicating that stress-induced APC/C activation is necessary for HT-induced mitotic slippage. Finally, HT significantly increased PTX cytotoxicity, regardless of cancer cells' sensitivity to PTX, and this activity was superior to the combination of PTX with pro-TAME. Our data suggested that forced mitotic exit of cells arrested in mitosis by anti-mitotic drugs, such as PTX, can be a more successful anticancer strategy than blocking mitotic exit by inactivation of the APC/C.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Antineoplásicos/farmacología , Hipertermia Inducida/métodos , Mitosis/fisiología , Neoplasias/tratamiento farmacológico , Paclitaxel/farmacología , Western Blotting , Ciclina B/metabolismo , Humanos , Mitosis/efectos de los fármacos
8.
Int J Oncol ; 42(6): 2069-77, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23591552

RESUMEN

Kaempferol belongs to the flavonoid family and has been used in traditional folk medicine. Here, we investigated the antitumor effects of kaempferol on cell cycle arrest and autophagic cell death in SK-HEP-1 human hepatic cancer cells. Kaempferol decreased cell viability as determined by MTT assays and induced a G2/M phase cell cycle arrest in a concentration-dependent manner. Kaempferol did not induce DNA fragmentation, apoptotic bodies or caspase-3 activity in SK-HEP-1 cells as determined by DNA gel electrophoresis, DAPI staining and caspase-3 activity assays, respectively. In contrast, kaempferol is involved in the autophagic process. Double-membrane vacuoles, lysosomal compartments, acidic vesicular organelles and cleavage of microtubule-associated protein 1 light chain 3 (LC3) were observed by transmission electron microscopy, LysoΤracker red staining, GFP-fluorescent LC3 assays and acridine orange staining, respectively. In SK-HEP-1 cells, kaempferol increased the protein levels of p-AMPK, LC3-II, Atg 5, Atg 7, Atg 12 and beclin 1 as well as inhibited the protein levels of CDK1, cyclin B, p-AKT and p-mTOR. Taken together, CDK1/cyclin B expression and the AMPK and AKT signaling pathways contributed to kaempferol-induced G2/M cell cycle arrest and autophagic cell death in SK-HEP-1 human hepatic cancer cells. These results suggest that kaempferol may be useful for long-term cancer prevention.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Ciclina B/metabolismo , Quempferoles/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
9.
J Acupunct Meridian Stud ; 6(2): 89-97, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23591004

RESUMEN

Although recent studies report that combined treatment of herbal drugs with acupuncture can improve clinical efficacy in traditional oriental medicine, experimental evidence that supports this pharmacopuncture therapy is rare thus far. Here, we investigated the effects of the herbal drug recipe Sciatica 5 (SCTA5) and acupuncture stimulation on gall bladder 30 (GB30) on regenerative responses of injured sciatic nerve in rats. Treatment of cultured dorsal root ganglion (DRG) neurons with SCTA5 improved neurite outgrowth. In vivo regenerative responses, in terms of distal extension of regenerating axons and retrogradely-labeled DRG neurons, were improved by either injury site application of SCTA5 or GB30 acupuncture stimulation and further increased by SCTA5 pharmacopuncture on GB30 acupoint. Moreover, combined treatment of SCTA5 and GB30 was more effective than singular treatments in inducing Cdc2 kinase and accompanying vimentin phosphorylation in Schwann cells of the injured nerve. These results suggest that SCTA5 and GB30 therapies may be cooperative in facilitating axonal regeneration in the injured peripheral nerves.


Asunto(s)
Terapia por Acupuntura/métodos , Regeneración Nerviosa/efectos de los fármacos , Extractos Vegetales/farmacología , Nervio Ciático/lesiones , Nervio Ciático/fisiología , Análisis de Varianza , Animales , Western Blotting , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Ciclina B/metabolismo , Ganglios Espinales/química , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Inmunohistoquímica , Masculino , Ratas , Ratas Sprague-Dawley , Nervio Ciático/citología
10.
Ann Bot ; 110(8): 1581-91, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23087128

RESUMEN

BACKGROUND AND AIMS: Prolonged treatment of Allium cepa root meristems with changing concentrations of hydroxyurea (HU) results in either premature chromosome condensation or cell nuclei with an uncommon form of biphasic chromatin organization. The aim of the current study was to assess conditions that compromise cell cycle checkpoints and convert DNA replication stress into an abnormal course of mitosis. METHODS: Interphase-mitotic (IM) cells showing gradual changes of chromatin condensation were obtained following continuous 72 h treatment of seedlings with 0·75 mm HU (without renewal of the medium). HU-treated root meristems were analysed using histochemical stainings (DNA-DAPI/Feulgen; starch-iodide and DAB staining for H(2)O(2) production), Western blotting [cyclin B-like (CBL) proteins] and immunochemistry (BrdU incorporation, detection of γ-H2AX and H3S10 phosphorylation). KEY RESULTS: Continuous treatment of onion seedlings with a low concentration of HU results in shorter root meristems, enhanced production of H(2)O(2), γ-phosphorylation of H2AX histones and accumulation of CBL proteins. HU-induced replication stress gives rise to axially elongated cells with half interphase/half mitotic structures (IM-cells) having both decondensed and condensed domains of chromatin. Long-term HU treatment results in cell nuclei resuming S phase with gradients of BrdU labelling. This suggests a polarized distribution of factors needed to re-initiate stalled replication forks. Furthermore, prolonged HU treatment extends both the relative time span and the spatial scale of H3S10 phosphorylation known in plants. CONCLUSIONS: The minimum cell length and a threshold level of accumulated CBL proteins are both determining factors by which the nucleus attains commitment to induce an asynchronous course of chromosome condensation. Replication stress-induced alterations in an orderly route of the cell cycle events probably reflect a considerable reprogramming of metabolic functions of chromatin combined with gradients of morphological changes spread along the nucleus.


Asunto(s)
Ciclo Celular/genética , Replicación del ADN/efectos de los fármacos , ADN de Plantas/genética , Cebollas/genética , Proteínas de Plantas/metabolismo , Núcleo Celular/genética , Tamaño de la Célula , Cromatina/genética , Ciclina B/metabolismo , Daño del ADN , Desoxirribonucleósidos/metabolismo , Técnica del Anticuerpo Fluorescente , Histonas/metabolismo , Peróxido de Hidrógeno/metabolismo , Hidroxiurea/farmacología , Meristema/efectos de los fármacos , Meristema/genética , Meristema/metabolismo , Meristema/ultraestructura , Cebollas/efectos de los fármacos , Cebollas/metabolismo , Cebollas/ultraestructura , Fosforilación , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/genética , Raíces de Plantas/metabolismo , Raíces de Plantas/ultraestructura , Plantones/efectos de los fármacos , Plantones/genética , Plantones/metabolismo , Plantones/ultraestructura
11.
Arch Pharm Res ; 35(7): 1251-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22864748

RESUMEN

Medicinal herbs are the preferred candidates for drug discovery against human diseases including cancer. The roots of Prismatomeris connata have been used in traditional herbal medicine to treat many health problems, particularly pneumoconiosis. This study was to test the anti-tumor activity of 3-hydroxy-1,5,6-trimethoxy-2-methyl-9,10-anthraquinone (PCON6), a major anthraquinone derivative from C. connata, against lung cancer. Cell viability in cultures was assessed by MTT assay. Cell death or apoptosis was determined with annexin-V and 7-aminoactinomycin D staining. Cell cycle was analyzed by both propidium iodide DNA staining and BrdU incorporation assay. Here we showed that in a panel of fifteen different tumor cells lines, a group of four non-small cell lung carcinoma (NSCLC) cell lines exhibited a relatively higher sensitivity to PCON6 growth inhibition than the rest of most non-lung cancer cell lines (p = 0.0461). Further studies demonstrated that the suppression of NSCLC H520 cell growth by PCON6 was associated with its induction of apoptosis at 20 µM (p = 0.0008), and of cell accumulation at S phase cell cycle (p < 0.05) that was further supported by a decrease in cdc2 protein expression. This preliminary study suggests that natural compound PCON6 has relatively selective cytotoxicity against NSCLC growth and represent a concept of developing a novel drug therapy specific for NSCLC based on the roots of C. connata or PCON6.


Asunto(s)
Antraquinonas/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2 , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Neoplasias Pulmonares/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos
12.
Planta Med ; 78(9): 890-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22532019

RESUMEN

Xanthatin, a natural bioactive compound of sesquiterpene lactones, was isolated and purified from air-dried aerial part of Xanthium sibiricum Patrin ex Widder. In the present study, we demonstrated the significant antiproliferative and proapoptotic effects of xanthatin on human gastric carcinoma MKN-45 cells. MTS assay showed that xanthatin produced obvious cytotoxicity in MKN-45 cells with IC50 values of 18.6, 9.3, and 3.9 µM for 12, 24, and 48 h, respectively. Results of flow cytometry analysis indicated that the antiproliferative activity induced by xanthatin might be executed via G2/M cell cycle arrest and proapoptosis in MKN-45 cells. Western blot analysis elucidated that: a) xanthatin downregulated expression of Chk1 and Chk2 and phosphorylation of CDC2, which are known as key G2/M transition regulators; b) xanthatin increased p53 activation, decreased the bcl-2/bax ratio and the levels of downstream procaspase-9 and procaspase-3, which are key regulators in the intrinsic apoptosis pathway; c) xanthatin blocked phosphorylation of NF-κB (p65 subunit) and of IκBα, which might contribute to its proapoptotic effects on MKN-45 cells. In conclusion, our results suggest that xanthatin may have therapeutic potential against human gastric carcinoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma/tratamiento farmacológico , Furanos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Antineoplásicos Fitogénicos/farmacología , Proteína Quinasa CDC2 , Carcinoma/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Ensayos de Selección de Medicamentos Antitumorales , Furanos/aislamiento & purificación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Genes p53 , Humanos , Proteínas I-kappa B/metabolismo , Concentración 50 Inhibidora , Inhibidor NF-kappaB alfa , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Neoplasias Gástricas/patología , Factor de Transcripción ReIA/metabolismo , Proteína X Asociada a bcl-2/metabolismo
13.
J Ethnopharmacol ; 141(3): 803-9, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22440953

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Acacia nilotica is widely distributed in Asia. In India, it occupies an important place in the indigenous system of medicine against anti-inflammatory, antioxidant, cancers, and/or tumors. AIM OF THE STUDY: The purpose of this study is to investigate the inhibitory effect of Acacia nilotica leaves extract and γ-Sitosterol on cell proliferation, the apoptotic effect and cell cycle arrest in breast and lung cancer cells. MATERIALS AND METHODS: GC-MS and HPLC were used to determine the chemical constituents of this extract and γ-Sitosterol respectively. Human MCF-7 and A549 cell lines were treated with Acacia nilotica extract and γ-Sitosterol. Cell viability was determined by MTT assay. Cell proliferation was determined by BrdU incorporation assay. Apoptosis was detected by cell morphologic observation through AO/EtBr staining, cell cycle analysis, and immunoblot analysis on the expression of protein associated with cell cycle arrest. RESULTS: Experimental results of bioactive compound analysis indicate that γ-Sitosterol, bioactive ingredients of Acacia nilotica extract. The IC(50) value of extract on MCF-7 and A549 cancer cells was 493.3±15.2 and 696.6±11.5 µg/ml, respectively. Acacia nilotica extract and γ-Sitosterol were inhibited the cell proliferation by 54.34±1.8 and 42.18±3.9% for MCF-7 and 58.26±1.5 and 44.36±3.05% for A549 cells. The percentage of apoptotic cells observed in the MCF-7 and A549 cell lines were increased to 42.46 and 36.8% of extract; 46.68 and 43.24% for γ-Sitosterol respectively. Flow cytometric analysis results demonstrate that cells were arrested at the G2/M phase and decrease the c-Myc expression. CONCLUSIONS: This study demonstrates in vitro results, which support the ethnomedical use of γ-Sitosterol against cancer. Experimental results of this study suggest that γ-Sitosterol exerts potential anticancer activity through the growth inhibition, cell cycle arrest and the apoptosis on cancer cells.


Asunto(s)
Acacia , Antineoplásicos Fitogénicos/farmacología , Extractos Vegetales/farmacología , Sitoesteroles/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B/metabolismo , Ciclina E/metabolismo , Humanos , Extractos Vegetales/química , Hojas de la Planta , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sitoesteroles/aislamiento & purificación
14.
J Neurooncol ; 107(2): 247-56, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22052333

RESUMEN

Malignant gliomas are a highly aggressive type of brain tumor with extremely poor prognosis. These tumors are highly invasive and are often surgically incurable and resistant to chemotherapeutics and radiotherapy. Thus, novel therapies that target pathways involved in growth and survival of the tumor cells are required for the treatment of this class of brain tumors. Previous studies revealed that epidermal growth factor receptor and extracellular-signal-regulated kinases (ERKs), which are involved in the induction of cell proliferation, are activated in the most aggressive type of glioma, i.e. glioblastoma multiforme (GBM). In fact, GBMs with increased levels of ERK activity exhibit a more aggressive phenotype than the others with moderate ERK activity, pointing to the importance of ERK and its kinase activity in the development and progression of these tumors. In this study, we have evaluated the effect of p38SJ, a novel member of the DING family of proteins, derived from Hypericum perforatum calluses, on the growth of malignant glioma cell lines, T98G and U-87MG by focusing on cell cycle and signaling pathways controlled by phosphorylation of various regulatory proteins including ERK. p38SJ, which exhibits profound phosphatase activity, shows the capacity to affect the phosphorylation status of several important kinases modulating signaling pathways, and cell growth and proliferation. Our results demonstrate that p38SJ reduces glioma cell viability and arrests cell cycle progression at G0/G1. The observed growth inhibitory effect of p38SJ is likely mediated by the downregulation of several cell cycle gatekeeper proteins, including cyclin E, Cdc2, and E2F-1. These results suggest that p38SJ may serve as a potential candidate for development of a therapeutic agent for the direct treatment of malignant gliomas and/or as a potential radiosensitizer.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/fisiopatología , Ubiquitina-Proteína Ligasas/farmacología , Animales , Proteína Quinasa CDC2 , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Transformada , Línea Celular Tumoral , Ciclina B/metabolismo , Ciclina E/metabolismo , Quinasas Ciclina-Dependientes , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Endopeptidasa K/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Glioblastoma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Hypericum/química , Ratones , Preparaciones de Plantas/farmacología , Complejo Represivo Polycomb 1 , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
Oncol Rep ; 27(3): 650-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22134635

RESUMEN

Recently, mild hyperthermia was shown to induce cell cycle arrest at the G2/M phase transition without leading to DNA damage. The mechanism of this regulation has not yet been elucidated, although p53 has been shown to be activated in response to mild hyperthermia. Here, we report the role of thioredoxin (TXN) in mild hyperthermia-induced cellular responses. Our data showed that the protein levels of p53 and its downstream gene, Gadd45a, which is an indicator of G2/M arrest, were significantly decreased in TXN siRNA-treated cells under conditions of mild hyperthermia (41˚C, 60 min) as compared to TXN wild-type cells, implying that TXN might play an important role in mild hyperthermia-induced G2/M arrest via p53 and Gadd45a activation. Furthermore, the release of cyclin-dependent kinase Cdc2, known to be regulated by Gadd45a under G2/M arrest, was inhibited from the nucleus for arrest in the G2/M phase in TXN downregulated cells under mild hyperthermia. We suggest that G2/M arrest mediated via the TXN-modulated p53 in response to mild hyperthermia may provide critical insight into the clinical use of mild hyperthermia to induce an adaptive response against genotoxic stresses.


Asunto(s)
Hipertermia Inducida , Tiorredoxinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína Quinasa CDC2 , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Ciclina B/genética , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Daño del ADN/genética , Regulación hacia Abajo , Fase G2/genética , Humanos , Mitosis/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Tiorredoxinas/genética , Proteína p53 Supresora de Tumor/genética
16.
Plant Cell ; 23(12): 4382-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22167058

RESUMEN

Increased cellular ploidy is widespread during developmental processes of multicellular organisms, especially in plants. Elevated ploidy levels are typically achieved either by endoreplication or endomitosis, which are often regarded as modified cell cycles that lack an M phase either entirely or partially. We identified GIGAS CELL1 (GIG1)/OMISSION OF SECOND DIVISION1 (OSD1) and established that mutation of this gene triggered ectopic endomitosis. On the other hand, it has been reported that a paralog of GIG1/OSD1, UV-INSENSITIVE4 (UVI4), negatively regulates endoreplication onset in Arabidopsis thaliana. We showed that GIG1/OSD1 and UVI4 encode novel plant-specific inhibitors of the anaphase-promoting complex/cyclosome (APC/C) ubiquitin ligase. These proteins physically interact with APC/C activators, CDC20/FZY and CDH1/FZR, in yeast two-hybrid assays. Overexpression of CDC20.1 and CCS52B/FZR3 differentially promoted ectopic endomitosis in gig1/osd1 and premature occurrence of endoreplication in uvi4. Our data suggest that GIG1/OSD1 and UVI4 may prevent an unscheduled increase in cellular ploidy by preferentially inhibiting APC/C(CDC20) and APC/C(FZR), respectively. Generation of cells with a mixed identity in gig1/osd1 further suggested that the APC/C may have an unexpected role for cell fate determination in addition to its role for proper mitotic progression.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Proteínas de Ciclo Celular/metabolismo , Mitosis , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Alelos , Ciclosoma-Complejo Promotor de la Anafase , Arabidopsis/citología , Arabidopsis/crecimiento & desarrollo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas Cdc20 , Proteínas de Ciclo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromosomas de las Plantas/genética , Cromosomas de las Plantas/metabolismo , Clonación Molecular , Cotiledón/genética , Cotiledón/metabolismo , Ciclina B/genética , Ciclina B/metabolismo , Citocinesis , Inhibidores Enzimáticos/metabolismo , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Prueba de Complementación Genética , Mutación , Células Vegetales/metabolismo , Epidermis de la Planta/citología , Epidermis de la Planta/genética , Epidermis de la Planta/crecimiento & desarrollo , Epidermis de la Planta/metabolismo , Estomas de Plantas/citología , Estomas de Plantas/genética , Estomas de Plantas/crecimiento & desarrollo , Estomas de Plantas/metabolismo , Plantas Modificadas Genéticamente/citología , Plantas Modificadas Genéticamente/genética , Plantas Modificadas Genéticamente/crecimiento & desarrollo , Plantas Modificadas Genéticamente/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Ploidias , Polen/crecimiento & desarrollo , Polen/metabolismo , Mapeo de Interacción de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos , Complejos de Ubiquitina-Proteína Ligasa/genética
17.
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
18.
Plant Cell ; 23(3): 1033-46, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21441434

RESUMEN

The anaphase-promoting complex/cyclosome (APC/C), an essential ubiquitin protein ligase, regulates mitotic progression and exit by enhancing degradation of cell cycle regulatory proteins, such as CYCB1;1, whose transcripts are upregulated by DUO POLLEN1 (DUO1). DUO1 is required for cell division in male gametophytes and is a target of microRNA 159 (miR159) in Arabidopsis thaliana. Whether APC/C is required for DUO1-dependent CYCB1;1 regulation is unknown. Mutants in both APC8 and APC13 had pleiotrophic phenotypes resembling those of mutants affecting microRNA biogenesis. We show that these apc/c mutants had reduced miR159 levels and increased DUO1 and CYCB1;1 transcript levels and that APC/C is required to recruit RNA polymerase II to MIR159 promoters. Thus, in addition to its role in degrading CYCB1;1, APC/C stimulates production of miR159, which downregulates DUO1 expression, leading to reduced CYCB1;1 transcription. Both MIR159 and APC8-yellow fluorescent protein accumulated in unicellular microspores and bicellular pollen but decreased in tricellular pollen, suggesting that spatial and temporal regulation of miR159 by APC/C ensures mitotic progression. Consistent with this, the percentage of mature pollen with no or single sperm-like cells increased in apc/c mutants and plants overexpressing APC8 partially mimicked the duo1 phenotype. Thus, APC/C is an integrator that regulates both microRNA-mediated transcriptional regulation of CYCB1;1 and degradation of CYCB1;1.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/genética , Ciclina B/metabolismo , MicroARNs/metabolismo , Polen/crecimiento & desarrollo , Complejos de Ubiquitina-Proteína Ligasa/genética , Secuencia de Aminoácidos , Ciclosoma-Complejo Promotor de la Anafase , Arabidopsis/citología , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Pleiotropía Genética , Mitosis , Datos de Secuencia Molecular , Fenotipo , Hojas de la Planta/genética , Hojas de la Planta/crecimiento & desarrollo , Plantas Modificadas Genéticamente/genética , Mutación Puntual , Polen/citología , Polen/metabolismo , ARN Polimerasa II/metabolismo , ARN de Planta/metabolismo , Elementos Reguladores de la Transcripción , Saccharomyces/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo
19.
Planta Med ; 77(10): 992-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21267808

RESUMEN

In this study, we incubated human A549 lung cancer cells with quercetin-metabolite-enriched plasma (QMP) obtained from Mongolian gerbils 2 h after quercetin feeding (100 mg/kg body wt/week). We investigated the effects of QMP on the growth of A549 cells and the possible mechanisms for these effects. We found that QMP but not control plasma (CP) reduced the cell growth in A549 cells. QMP led to cell cycle arrest at the G (2)/M phase by downregulating the expression of cdk1 and cyclin B. QMP but not CP or quercetin itself significantly increased PPAR- γ expression (p < 0.05), which was accompanied by an increase of phosphatase and tensin homologue deleted on the chromosome ten level and a decrease of phosphorylation of Akt. Furthermore, quercetin-3-glucuronide and quercetin-3'-sulfate also significantly increased PPAR- γ expression in A549 cells. GW9662, a PPAR- γ antagonist, significantly suppressed the effects of 10 % QMP on cell proliferation and on the expression of cyclin B and cdk1. Taken together, these data suggest that the activation of PPAR- γ plays an important role, at least in part, in the antiproliferative effects of quercetin metabolites.


Asunto(s)
División Celular/efectos de los fármacos , Fase G2/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , PPAR gamma/metabolismo , Quercetina/metabolismo , Quercetina/farmacología , Administración Oral , Anilidas/farmacología , Animales , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Gerbillinae , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Proteína Oncogénica v-akt/efectos de los fármacos , Proteína Oncogénica v-akt/metabolismo , PPAR gamma/antagonistas & inhibidores , Fosfohidrolasa PTEN/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Plasma/metabolismo , Quercetina/administración & dosificación , Quercetina/análogos & derivados , Regulación hacia Arriba/efectos de los fármacos
20.
Int J Oncol ; 37(2): 493-501, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20596677

RESUMEN

Mitotane inhibits steroid synthesis by an action on steroidogenic enzymes, as 11beta-hydroxylase and cholesterol side chain cleavage. It also has a cytotoxic effect on the adrenocortical cells and represents a primary drug used in the adrenocortical carcinoma (ACC). H295R and SW13 cell lines were treated with mitotane 10(-5) M and ionizing radiations (IR) in combination therapy, inducing an irreversible inhibition of cell growth in both adrenocortical cancer cells. As shown in a previous report, mitotane/IR combination treatment induced a cell accumulation in the G2 phase. Here, we report the radiosensitizing properties of mitotane in two different ACC cell lines. The drug reveals the effectiveness to enhance the cytotoxic effects of IR by attenuating DNA repair and interfering on the activation of mitosis promoting factor (MPF), mainly regulated by the degradation of cyclin B1 in the mitotic process. These events may explain the inappropriate activation of cdc2, implicated in G2/M phase arrest and probably induced by the mitotane and IR in the combined treatment. Indeed, treatment with purvalanol, a cdc2-inhibitor prevents cell cycle arrest, triggering the G2/M transition. The observation that mitotane and IR in combination treatment amplifies the activation level of cyclin B/cdc2 complexes contributing to cell cycle arrest, suggests that the MPF could function as a master signal for controlling the temporal order of different mitotic events. Moreover, we report that mitotane interferes in modulation of mismatch repair (MMR) enzymes, revealing radiosensitizing drug ability.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/radioterapia , Carcinoma Corticosuprarrenal/radioterapia , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Mitotano/farmacología , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Reparación de la Incompatibilidad de ADN/efectos de los fármacos , Reparación de la Incompatibilidad de ADN/fisiología , Evaluación Preclínica de Medicamentos , Fase G2/efectos de los fármacos , Fase G2/fisiología , Humanos , Complejos Multiproteicos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Radiación Ionizante , Fármacos Sensibilizantes a Radiaciones/farmacología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA