Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Yao Xue Xue Bao ; 51(2): 309-15, 2016 02.
Artículo en Zh | MEDLINE | ID: mdl-29856586

RESUMEN

This study was designed to investigate the effect of Xiao-Ai-Ping injection on cancer angiogenesis. CCK8 assay and Brd U incorporation immunofluorescence assay were used to detect the effect of Xiao-Ai-Ping injection on HUVECs proliferation; wound healing assay and transwell assay were employed to test the effect of Xiao-Ai-Ping injection on HUVECs migration. The anti-angiogenic effect of Xiao-Ai-Ping injection was examined by tube formation assay, rat aortic ring assay and chicken chorioallantoic membrane(CAM) assay. ELISA assay was used to measure the secretion of vascular endothelial growth factor(VEGF); and the activation of vascular endothelial growth factor receptor 2(VEGFR2) protein and its downstream signaling pathways were examined by Western blot. Our data demonstrated that Xiao-Ai-Ping injection inhibited HUVECs proliferation in a time- and dose-dependent manner, and the IC(50) (mg·m L(-1)) values for 24, 48 and 72 h were 48.7 ± 7.14, 29.1 ±2.25 and 22.0 ± 4.53, individually. Xiao-Ai-Ping injection inhibited HUVECs DNA synthesis and migration. Xiao-Ai-Ping injection suppressed HUVECs tube formation, and reduced microvessel sprouting from rat aortic rings and vessel growth in CAMs. Furthermore, Xiao-Ai-Ping injection attenuated the secretion of VEGF, and inhibited the expression of p-VEGFR2 and phosphorylation of protein kinase B(p-AKT). We conclude that Xiao-Ai-Ping injection inhibits angiogenesis by down-regulation of VEGF signaling and AKT pathway.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Medicamentos Herbarios Chinos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Animales , Movimiento Celular , Pollos , Membrana Corioalantoides , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
2.
Bioorg Med Chem Lett ; 25(9): 1976-8, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25817591

RESUMEN

(+)- and (-)-liriodenol, a pair of unprecedented enantiomeric lignans bearing a 1,1-disubstituted olefinic group, were isolated from the barks of Liriodendron hybrid. The structure and relative configurations were determined by comprehensive analysis of MS and NMR spectroscopy. The cytotoxicity of these three lignans ((±)-, (+)-, and (-)-liriodenol) was evaluated in vitro against four selected human tumor cell lines, where (+)-liriodenol showed more significant cytotoxic effects than the (±)- and (-)-liriodenol enantiomers.


Asunto(s)
Lignanos/química , Lignanos/farmacología , Liriodendron/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colorimetría , Relación Dosis-Respuesta a Droga , Humanos , Lignanos/aislamiento & purificación , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad
3.
Z Naturforsch C J Biosci ; 70(11-12): 305-11, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26618570

RESUMEN

The effect of LXB-1, an extract from Liriodendron × hybrid, was determined on A549 human lung adenocarcinoma cell lines. Growth inhibition of LXB-1 was analyzed by MTT assay. Cancer cell cycle was measured by flow cytometry. To verify the apoptosis effect of LXB-1 on A549 cells, annexin V/PI double staining assay was performed. The expression levels of proapoptotic proteins were also measured by western blot. The potential mechanisms of LXB-1 inducing apoptosis - the expression and phosphorylation of ERK, p38, JNK and Akt - were investigated by western blot. The IC50 values of LXB-1 on A549 for 24, 48 and 72 h treatment were determined to be 12.97±1.53 µg/mL, 9.55±1.42 µg/mL, and 5.90±0.74 µg/mL, respectively. LXB-1 induced an obvious G2/M cell cycle arrest in A549 cells and resulted in significant cell apoptosis. LXB-1 also increased the cleavage of both caspase-3 and caspase-9, and greatly decreased the protein levels of Bcl-2. Moreover, LXB-1 increased the expression of phosphorylated JNK but decreased the levels of phosphorylated ERK1/2 and Akt. These results suggest that that LXB-1 induced apoptosis through JNK, ERK1/2, and Akt pathways in A549 cells.

4.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 56-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24274976

RESUMEN

In this study, we investigated the role and molecular mechanism of p43 and YS-1 (recombinant human p43 protein) in Dll4-Notch1 signaling pathway. Active, small interfering RNA and recombinant plasmid targeting of p43 protein were used to infect human umbilical vein endothelial cells (HUVECs). Three-dimensional sprouting model, endothelial cell migration assay, and sprouting and tube formation assay were used to deduce the function of p43 and YS-1 in angiogenesis. Semi-quantitative reverse transcription-polymerase chain reaction and western blot analysis were performed to detect the efficiency of p43 in Dll4-Notch1 signaling in HUVECs. It was found that silencing and overexpression of p43 could upregulate Dll4-Notch and stimulate angiogenesis. p43 plays a complex role in angiogenesis. When the concentration is under 100 nM, it promotes angiogenesis; instead, when the concentration is over 100 nM, it inhibits angiogenesis. In this study, we found that the expression level of p43 was under 60 nM. However, recombinant human p43 protein, YS-1, inhibited endothelial cell sprouting, and 500 µg/ml of YS-1 attenuated the activation of Dll4-Notch1 signaling. These results suggested that YS-1 could directly inhibit angiogenesis through Dll4-Notch1 signal transduction pathway, while p43 plays a modulating role in this signaling pathway.


Asunto(s)
Proteínas Mitocondriales/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Factor Tu de Elongación Peptídica/farmacología , Receptor Notch1/fisiología , Transducción de Señal/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/fisiopatología , Proteínas Mitocondriales/biosíntesis , Factor Tu de Elongación Peptídica/biosíntesis , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/farmacología , Factor A de Crecimiento Endotelial Vascular/fisiología
5.
Acta Oncol ; 52(4): 842-51, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23409769

RESUMEN

BACKGROUND: Hypoxia up-regulated expression of tissue factor (TF) may facilitate tumor cell metastasis, but transcriptional mechanisms remain undefined. MATERIAL AND METHODS: To verify the role of Egr-1 in hypoxia-induced TF expression in breast cancer cells, quantitative PCR and Western blot analysis were performed. The secretion of VEGF under hypoxia was detected by enzyme-linked immunosorbent assay (ELISA). Egr-1 and HIF-1α siRNA were transiently transfected into breast cancer cells to evaluate their specific roles. RESULTS: The increased Egr-1 expression occurring in hypoxic breast cancer cells can up-regulate TF expression and stimulating protein 1(Sp1) was not responsible for the hypoxia-induced expression of TF. HIF-1α mediated the hypoxia-induced up-regulation of TF expression through vascular endothelial growth factor (VEGF). The regulatory effects of Egr-1 on TF under hypoxia were independent of HIF-1α. Either Egr-1 or HIF-1α was responsible for hypoxic induction of tumor cells adhesion. HIF-1α, but not Egr-1, had a pivotal role in human breast cancer cells invasion. Both Egr-1 and HIF-1α were critical to angiogenesis induced by hypoxic conditions in MDA-MB-231 and HUVEC co-cultures. In nude mice, both Egr-1 and HIF-1α small interfering RNA (siRNA) could decrease extravasation of MDA-MB-435 cells in the lung after tail vein injection. CONCLUSIONS: Hypoxia-induced expression of TF in human breast cancer cells depends on Egr-1 and HIF-1α, and both of these proteins may play an important role in breast cancer metastasis, either directly or indirectly through the TF pathway.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Tromboplastina/genética , Animales , Neoplasias de la Mama/genética , Carcinoma/genética , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Células Cultivadas , Técnicas de Cocultivo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Tromboplastina/metabolismo
6.
Cancer Sci ; 103(5): 904-12, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22380870

RESUMEN

The chemokine CC motif receptor 5 (CCR5) and its ligands have been reported to be associated with cancer progression and metastasis. Although recent researches have demonstrated a fundamental role of hypoxia in cancer, the effect of hypoxia on the expression and function of CCR5 and its ligands in cancer cells is unknown. Here, we investigated the status of CCR5 and its ligands in cancer cells under hypoxic conditions. Quantitative polymerase chain reaction, western blotting and immunofluorescence staining showed that hypoxia induced a strong increase of CCR5 expression. Dual luciferase assay and mRNA stability analysis indicated that hypoxia-induced CCR5 mRNA expression relied on both transcriptional and posttranscriptional mechanisms. We detected the expression of CCR5 ligands and found that chemokine CC motif ligand 5 (CCL5) was induced under hypoxia. Recombinant human CCL5 stimulated cell migration rather than cell proliferation under hypoxia, and neutralization of CCL5 inhibited hypoxia-induced migration of cancer cells. Similarly, overexpression of CCR5 increased cell migration, and knockdown of CCR5 attenuated hypoxia-mediated cell migration. We further showed that hypoxia-inducible factor-1α (HIF-1α) was involved in CCR5 and CCL5 regulation under hypoxia. HIF-1α mRNA levels were highly correlated with CCR5 mRNA and CCL5 mRNA levels in clinical samples. CCR5 and CCL5 were highly expressed in breast cancer lymph nodes metastases. Taken together, our data suggest that CCR5-CCL5 interaction promotes cancer cell migration under hypoxia.


Asunto(s)
Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Movimiento Celular , Quimiocina CCL5/metabolismo , Receptores CCR5/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metástasis Linfática , Persona de Mediana Edad , Interferencia de ARN , Receptores CCR5/genética , Transfección
7.
Biol Pharm Bull ; 33(7): 1192-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606312

RESUMEN

Adhesion and migration of tumor cells are crucial steps in tumor invasion and metastasis. In the present study, we investigated the effects of saponin monomer 13 of dwarf lilyturf tuber (DT-13) on metastasis of human breast cancer cells (MDA-MB-435) during hypoxia. The effects and molecular mechanisms of DT-13 on MDA-MB-435 cells metastatic phenotype in vitro and in vivo were evaluated by RNA interference; quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays. DT-13 had no significant effects on cell adhesion and migration under normoxia conditions. Under hypoxic conditions, MDA-MB-435 adhesion to vitronectin was inhibited by about 43.5% or 60.8% after exposure of the cells to DT-13 at 1 microM or 10 microM, respectively. DT-13 decreased the migratory response by hypoxia at 1 or 10 microM, and inhibition ratios were 20% and 30%, respectively. DT-13 inhibited hypoxia-induced expression of alphavbeta3 integrin, tissue factor (TF) and early growth response gene-1 (Egr-1) and decreased excretion of matrix metalloproteinase 9 (MMP-9) of MDA-MB-435 cells under hypoxic conditions. After Egr-1 short interference RNA (siRNA) treatment, DT-13 could still inhibit the up-regulation of TF mRNA and protein levels and its pro-coagulant activity (PCA) under hypoxia. In nude mice, DT-13 decreased extravasation of MDA-MB-435 cells in the lung after tail vein injection. Our data suggest that DT-13 inhibits MDA-MB-435 cells metastasis during hypoxia via regulation of TF, and the effect of DT-13 on TF is partly mediated by Egr-1.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Hipoxia de la Célula , Movimiento Celular/efectos de los fármacos , Liriope (Planta)/química , Saponinas/farmacología , Tromboplastina/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Acta Pharmacol Sin ; 30(5): 628-36, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19363519

RESUMEN

AIM: The aim of this study was to investigate the effect of topotecan (TPT) on cancer cell migration. METHODS: Growth inhibition of TPT was analyzed by MTT assay, and cancer cell migration was measured by transwell double chamber assay. To verify the effect of TPT on the chemokine receptors CXCR4 and CCR7, quantitative PCR, semi-quantitative PCR and Western blot analysis were performed. The secretion of MMP-2 and MMP-9 was detected by enzyme-linked immunosorbent assay (ELISA) and gelatin zymography. To evaluate possible contributions of CCR7 to MMP secretion, the overexpression vectors pcDNA3.1(+)-CCR7 and CCR7 siRNA were transiently transfected into MDA-MB-435 cells. RESULTS: TPT inhibited cancer cell migration in a dose-dependent manner. Additionally, TPT significantly decreased the expression of CCR7 in both MDA-MB-435 and MDA-MB-231 cells and moderately reduced the expression of CXCR4 in MDA-MB-435 cells. The secretion of MMPs (MMP-2, MMP-9) was also inhibited by TPT. Overexpression of CCR7 increased the secretion of MMP-2/9 and cancer cell migration, whereas knockdown of CCR7 reduced active MMP-2/9 production and migration of MDA-MB-435 cells. CONCLUSION: TPT inhibited cancer cell migration by down-regulation of CCR7 and MMPs (MMP-2 and MMP-9).


Asunto(s)
Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores CCR7/metabolismo , Topotecan/farmacología , Línea Celular Transformada , Línea Celular Tumoral , Expresión Génica/efectos de los fármacos , Humanos , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Receptores CCR7/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Topotecan/uso terapéutico , Regulación hacia Arriba
9.
Onco Targets Ther ; 12: 10811-10825, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31853183

RESUMEN

BACKGROUND: Camellia nitidissima Chi (CNC) has been applied as a traditional folk medicine for the effective treatment of various diseases. However, there is little research regarding the mechanism of CNC on pharmaceutical function including anticancer effect. MATERIALS AND METHODS: JHC-4 is a n-butanol extract from CNC. The anti-proliferation effect was evaluated by MTT assays. Monodansylcadaverine (MDC) staining, Western blotting and autophagy inhibitors (CQ and BafA1) were applied to determine whether JHC-4 induced autophagy. The synergistic anticancer effect was evaluated by MTT assays, flow cytometry, Western blotting and autophagy inhibitors. Western blotting was used to explore the influence of PI3K/Akt/mTOR signaling pathway induced by drug treatment. RESULTS: JHC-4 caused significant growth inhibition and induced autophagy in human gastric cancer cells. Moreover, JHC-4 as an autophagy agonist synergistically potentiated the sensitivity of gastric cancer cells to paclitaxel. Meanwhile, JHC-4 could significantly enhance the growth inhibition effect of paclitaxel by the induction of autophagy and apoptosis. Finally, we demonstrated that the PI3K/Akt/mTOR signaling pathway was involved in the synergistic anti-proliferation effect of JHC-4 and paclitaxel. CONCLUSION: All these data indicated that JHC-4 was a novel autophagy inducer when combination with paclitaxel for gastric cancer, which provided the scientific evidence for the use of this Chinese traditional medicine against cancer.

10.
Eur J Pharmacol ; 818: 124-131, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29037767

RESUMEN

Natural medicine has multi-levels, multi-paths and multi-targets, and an increasing number of reports have confirmed that the combination of natural medicine with chemotherapy drugs exhibit a significant synergistic effect. It is necessary to find drug combination strategies to enhance efficacy and reduce toxicity, which can relieve the restrictions on the use of several chemotherapy drugs that have serious toxicity. Our previous reports showed that DT-13 inhibits cancer proliferation, invasion, migration, metastasis, and angiogenesis and induces autophagy. In this study, we evaluated the anti-proliferation effect of DT-13 on a panel of 40 different cancer cell lines for the first time. Moreover, it is also the first time that the combination of DT-13 with 5 different chemotherapy drugs on 3 common cancer cells has been examined. We further confirmed that DT-13 enhanced the sensitivity of gastric cancer cells to topotecan (TPT) via cell cycle arrest in vitro and in vivo. Considering that TPT has been subjected to restriction because of its serious toxicity, DT-13 showed the ability to enhance its effect and reduce its toxicity, which could provide a strategy to reduce the toxic and clinical side effects of TPT.


Asunto(s)
Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Saponinas/farmacología , Neoplasias Gástricas/patología , Topotecan/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos
11.
Int J Mol Med ; 41(5): 2793-2801, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29484370

RESUMEN

Chemotherapy is the preferred and most common treatment for cancer in clinical practice. An increasing number of researchers all over the world are focusing on natural medicines to find new antitumor drugs, and several reports have shown that Camellia nitidissima (C. nitidissima) Chi could reduce blood-lipid, decrease blood pressure, resist oxidation, prevent carcinogenesis and inhibit tumors. Therefore, the pharmacodynamics of the chemical constituents in C. nitidissima need to be investigated further. In the present study, 16 chemical constituents were isolated from the leaves of C. nitidissima, of which 6 compounds are reported to be found in this plant for the first time. Furthermore, all these phytochemicals were screened for antitumor activity on 4 common cancer cell lines, while compound 3, one oleanane-type triterpene, exhibited the most potential antitumor effects. Interestingly, to our knowledge, this was the first report that compound 3 inhibits cancer cells. Compound 3 inhibited EGFR-mutant lung cancer cell line, NCI-H1975 via apoptosis effect, with an IC50 of 13.37±2.05 µM at 48 h. Based on the data, compound 3 showed potential for antitumor drug development, suggesting the scientific basis for the antitumor activity of C. nitidissima.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Camellia/química , Neoplasias/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Extractos Vegetales/química , Extractos Vegetales/farmacología
12.
Oncol Rep ; 37(3): 1756-1764, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28098907

RESUMEN

Adriamycin (ADM) is a principal drug for the treatment of renal cell cancer (RCC). Due to its limited response and high renal and cardiac toxicity, synergistic effects of ADM in combination with other drugs have been widely researched. In this study, we found the combination between YS-1 and ADM, performed higher anticancer activity on 786-O human RCC cells in vitro and in vivo, than that reported on its anti-angiogenesis effect compared with monotherapy of ADM. Our data showed that when combined with ADM, YS-1 promoted the sensitivity of 786-O cells to ADM. The combination of YS-1 and ADM also inhibited cell proliferation, but without affecting cell apoptosis. We found that ADM monotherapy treatment notably upregulated the activity of extracellular signal-regulated kinase ERK1 and ERK2 (ERK1/2), but when combined with YS-1, the p-ERK1/2 level was reduced; then inhibited the Ras/Raf/MEK pathway. Additionally, the synergistic effects on cell cycle arrest inhibition were eliminated when ERK1/2 was silenced using siRNA. Our combination therapy of YS-1 with ADM showed the strongest antitumor effects in vivo (inhibition ratio: 5 mg/kg YS-1 combined with 1 mg/kg ADM, 68.19%) in comparison with individual effects (inhibition ratio: 5 mg/kg YS-1, 30.07%; 1 mg/kg ADM, 50.42%). Collectively, these findings indicated that YS-1 did not only enhance the ability of ADM to inhibit tumor proliferation, but also reduce the renal toxicity to protect the normal renal tissues.


Asunto(s)
Doxorrubicina/farmacología , Sinergismo Farmacológico , Neoplasias Renales/prevención & control , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Mitocondriales/farmacología , Factor Tu de Elongación Peptídica/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Femenino , Humanos , Técnicas In Vitro , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biomed Pharmacother ; 89: 1277-1285, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28320095

RESUMEN

Vinorelbine (NVB) is a semi-synthetic vinca alkaloid that is approved for the clinical therapy of lung cancer. However, the clinical application of NVB was limited because of the acquisition of resistance and inacceptable toxicity. Therefore, it is of great interest to develop low-cytotoxic drugs that can synergize with NVB. DT-13, a saponin monomer 13 of the Dwarf lilyturf tuber, showed inhibitory effects on tumor metastasis and angiogenesis in the previous studies. Here, we found that DT-13 combined with NVB exhibited synergistic effect to inhibit the cell proliferation in human lung cancer NCI-H1299 cells rather than human embryonic lung fibroblasts WI-38. The combination of DT-13 and NVB significantly inhibited the colony formation, induced cellular and nuclear morphological changes, and triggered cell cycle arrest at mitotic phase. Furthermore, MAPK signaling pathway was activated by the combination treatment, and the activation of ERK was required for the induction of mitotic arrest. Taken together, DT-13 combined with NVB exhibited synergistic anticancer effect in NCI-H1299 cells, and DT-13 may be a candidate agent for adjuvant chemotherapy of NVB in lung cancer.


Asunto(s)
Liriope (Planta)/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mitosis/efectos de los fármacos , Saponinas/farmacología , Transducción de Señal/efectos de los fármacos , Vinblastina/análogos & derivados , Ciclo Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Vinblastina/farmacología , Vinorelbina
14.
Biomed Pharmacother ; 89: 805-811, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28273642

RESUMEN

Particular mutations in the isocitrate dehydrogenase gene (IDH) were discovered in several gliomas citing astrocytoma, oligodendroglioma, and glioblastoma multiform, but also in leukemia; these mutations were discovered in nearly all cases of secondary glioblastomas, they evolve from lower-grade gliomas, but are limited in primary high-grade glioblastoma multiform. These mutations distinctively produce (D)-2-hydroxyglutarate (D-2-HG) from alpha-ketoglutarate (α-KG). (D)-2-hydroxyglutarate is accumulated to very high concentrations which inhibit the function of enzymes that are dependent on alpha-ketoglutarate. This modification leads to a hyper-methylated state of DNA and histones, resulting in different gene expression that can activate oncogenes and inactivate tumor-suppressor genes. In our work we review the impact of the mutations that occur in IDH genes, we focus on their impact on distribution in cancer. As IDH mutations appear in many different conditions we expose the extent of IDH mutations and derivate their impact on cancer prognosis, diagnosis, and even their oncogenicity, we will also link their impact to HIF-1α and derivate some target and finally, we present some of the therapeutics under research and out on market.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Neoplasias/metabolismo , Biomarcadores , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isocitrato Deshidrogenasa/genética , Mutación , Neoplasias/genética
15.
Anticancer Agents Med Chem ; 17(11): 1500-1507, 2017 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-28356014

RESUMEN

BACKGROUND: The epidermal growth factor receptor (EGFR) is a driver oncogene and specific blockade of EGFR has been shown to be an effective therapeutic approach against multiple human cancers. AIMS: Here we employed the homogeneous time-resolved fluorescence (HTRF) technology to screen new EGFR mediators. METHOD: 4 hits (NDS-41107, NDS-41119, NDS-41111 and NDS-41126) were confirmed in a compound library of 8000 compounds, and the IC50 values were determined to be 15.45±2.25µM (NDS-41107), 6.16±0.88 µM (NDS-41119), 11.33±3.31 µM (NDS-41111) and 11.19±1.67µM (NDS-41126), respectively. We then showed that NDS-41119 (N-cyclohexyl-2-(1-(phenylsulfonyl) piperidin-4-yl) acetamide) significantly inhibited EGFR signaling in human lung cancer cells, as evidenced by decreased phosphorylation of EGFR、ERK and Akt. NDS-41119 also attenuated EGF-induced cell proliferation and migration in a dose-dependent manner. We finally demonstrated that NDS-41119 inhibited the T790M mutation in NCI-H1975 cells and potentiated the effect of gefitinib against resistant cells. RESULT: Our results will contribute to the development of novel EGFR-targeted anti-cancer drugs.


Asunto(s)
Acetamidas/farmacología , Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Transferencia Resonante de Energía de Fluorescencia , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Acetamidas/síntesis química , Acetamidas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química
16.
Cell Death Dis ; 8(5): e2810, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28542137

RESUMEN

Non-small-cell lung cancer (NSCLC) is the most commonly diagnosed malignant disease with the leading cause of cancer-related death. Combination treatment remains the major strategy in the clinical therapy of NSCLC. Vinorelbine (NVB), a semi-synthetic vinca alkaloid, is used for advanced and metastatic NSCLC by destabilizing microtubule formation to induce mitotic arrest and cell death. However, the side effect of NVB heavily affected its effectiveness in clinical therapy. Hence, it is of great significance to develop new agents to synergize with NVB and decrease the adverse effect. In our study, we found that the saponin monomer 13 of the dwarf lilyturf tuber, DT-13, exhibiting anti-angiogenesis and anti-metastasis effect, synergized with NVB to inhibit cell proliferation in NSCLC cells. The synergistic interaction of DT-13 and NVB was confirmed by combination Index values. Also, DT-13 and NVB act in concert to inhibit the long-term colony formation. Furthermore, DT-13/NVB co-treatment cooperated to induce mitotic arrest and subsequent apoptosis. Mechanistically, we found that nuclear expression of transcription factors forkhead box M1 (FOXM1) and levels of motor adaptor bicaudal D2 (BICD2) were dramatically reduced by combination treatment. Importantly, oncogene FOXM1 was identified as the crucial regulator of BICD2, which played critical roles in NVB-induced mitotic spindle defects. Moreover, overexpression of FOXM1 and BICD2 significantly reversed mitotic arrest induced by DT-13/NVB co-treatment, and siRNAs against both genes greatly increased the combinational effects. In addition, in vivo study revealed that DT-13 combined with NVB significantly suppressed tumor growth in nude mice xenograft model, and downregulated the expression of FOXM1 and BICD2 in tumor tissues, which was consistent with in vitro study. In conclusion, DT-13 might provide a novel strategy for the chemosensitization of NVB in NSCLC therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias Pulmonares/patología , Mitosis/efectos de los fármacos , Saponinas/farmacología , Vinblastina/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Proteína Forkhead Box M1/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Asociadas a Microtúbulos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Vinblastina/farmacología , Vinorelbina
17.
Cell Death Dis ; 8(10): e3143, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29072684

RESUMEN

In multiple types of tumors, fibrotic collagen is regarded as the 'highway' for cancer cell migration, which is mainly modified by lysyl hydroxylase 2 (PLOD2). The previous findings have demonstrated that the expression of PLOD2 was regulated by multiple factors, including HIF-1α, TGF-ß and microRNA-26a/b. Although PLOD2 was confirmed to be related to poor prognosis in lung adenocarcinoma, the regulatory mechanism and function of PLOD2 in human lung adenocarcinoma is poorly understood. On the other hand, upregulation or hyperactivation of epidermal growth factor receptor is considered as a prognostic marker in many cancers, especially in non-small-cell lung cancer (NSCLC). In this study, we found that PLOD2 was elevated in NSCLC specimens and positively links to NSCLC poor prognosis. Gain- and loss-of-function studies and orthotopic implantation metastasis model pinpointed that PLOD2 promotes NSCLC metastasis directly by enhancing migration and indirectly by inducing collagen reorganization. In addition, we revealed that PLOD2 was regulated by PI3K/AKT-FOXA1 axis. The transcription factor FOXA1 directly bound to the PLOD2 promoter, and turned on PLOD2 transcription. In summary, our findings revealed a regulatory mechanism of NSCLC metastasis through EGFR-PI3K/AKT-FOXA1-PLOD2 pathway, and provided PLOD2 as a therapeutic target for NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Neoplasias Pulmonares/metabolismo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Análisis de Supervivencia , Transfección
18.
Oncotarget ; 8(66): 110426-110443, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29299159

RESUMEN

Aberrant energy metabolism is critical for cancer progression. Tumor-associated macrophages (TAMs) can stimulate tumor angiogenesis and enhance cancer metastasis; however, the metabolic interaction between cancer cells and macrophages characterized by lactate shuttles remains unclear. Here, we showed that lactate activated human macrophages to a TAM-like phenotype and stimulated the secretion of CCL5 by activation of Notch signaling in macrophages. Reciprocally, CCL5 increased cell migration, induced cancer cell EMT, and promoted aerobic glycolysis in breast cancer cells, suggesting a positive metabolic feedback loop in the co-culture system. Inhibition of CCR5, the cognate receptor of CCL5, or neutralization of CCL5, broke the metabolic loop and decreased cancer cell migration and EMT. Inhibition of aerobic glycolysis significantly reduced breast cancer cell EMT, indicated that aerobic glycolysis was necessary for the invasive phenotype of cancer cells. We further showed that TGF-ß signaling regulated the expression of CCR5 in the co-culture system, and CCL5 induced glycolysis by mediation of AMPK signaling. The expression of CCL5-CCR5 axis was highly associated with macrophage infiltration, TGF-ß and p-AMPK in clinical samples. CCL5-CCR5 axis promoted breast cancer metastasis in vivo. Our findings suggested a pivotal role of CCL5-CCR5 axis in the metabolic communication between cancer cells and macrophages.

19.
Front Pharmacol ; 8: 447, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744216

RESUMEN

Colorectal cancer (CRC) is one of the most common malignant tumors worldwide, occurring in the colon or rectum portion of large intestine. With marked antioxidant, anti-inflammation and anti-tumor activities, Camellia nitidissima Chi has been used as an effective treatment of cancer. The azoxymethane/dextran sodium sulfate (AOM/DSS) induced CRC mice model was established and the prevention effect of C. nitidissima Chi extracts on the evolving of CRC was evaluated by examination of neoplastic lesions, histopathological inspection, serum biochemistry analysis, combined with nuclear magnetic resonance (NMR)-based metabolomics and correlation network analysis. C. nitidissima Chi extracts could significantly inhibit AOM/DSS induced CRC, relieve the colonic pathology of inflammation and ameliorate the serum biochemistry, and could significantly reverse the disturbed metabolic profiling toward the normal state. Moreover, the butanol fraction showed a better efficacy than the water-soluble fraction of C. nitidissima Chi. Further development of C. nitidissima Chi extracts as a potent CRC inhibitor was warranted.

20.
Cancer Chemother Pharmacol ; 58(4): 434-43, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16470410

RESUMEN

OBJECTIVES: To investigate the effects and potential mechanisms of gambogic acid (GA), a naturally occurring anticancer agent, on the expression and regulation of telomerase in human gastric carcinoma cells. METHODS: GA-induced inhibition of cell proliferation was evaluated by the commonly employed MTT assay on two human gastric carcinoma cell lines, MGC-803 and SGC-7901. Telomerase activity and hTERT mRNA expression were determined by telomeric repeat amplication protocol-polymerase chain reaction and reverse transcription-polymerase chain reaction, respectively. The hTERT promoter activity was measured by luciferase assay. The expression of c-MYC, an apoptotic gene that modulates the expression of hTERT promoter, was quantified by Western blotting. RESULTS: The proliferation of human gastric carcinoma cell lines, MGC-803 and SGC-7901, was significantly inhibited with GA treatment. Both telomerase activity and hTERT mRNA expression were notably decreased in cells treated with GA. The activity of hTERT promoter and the expression of c-MYC were also remarkably decreased in GA-treated cells. CONCLUSION: This study demonstrated that GA treatment of human gastric carcinoma cell lines, MGC-803 and SGC-7901, significantly reduced the expression of c-MYC in a time- and concentration-dependent manner accompanied with the down-regulation of the hTERT transcription and the ultimate reduction in telomerase activity. Our results indicate that the hTERT is a target of c-MYC activity and identify a feasible mechanism of GA's potent anticancer activity.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Fragmentos de Péptidos/efectos de los fármacos , Telomerasa/efectos de los fármacos , Xantonas/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Humanos , Fragmentos de Péptidos/metabolismo , ARN Mensajero , Neoplasias Gástricas/enzimología , Telomerasa/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA