Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Altern Ther Health Med ; 21 Suppl 2: 46-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26308760

RESUMEN

CONTEXT: Turmeric (Curcuma longa) is a food spice and colorant reported to be beneficial for human health. Curcumin (diferuloylmethane) is the major ingredient in turmeric, and existing data suggest that the spice, in combination with chemotherapy, provides a superior strategy for treatment of gastrointestinal cancer. However, despite its significant effects, curcumin suffers from poor bioavailability, due to poor absorption in the body. OBJECTIVE: The research team intended to evaluate a liquid extract of turmeric roots (TEx) that the team had formulated for its in vitro, anticancer activity against several human, colorectal cancer cell lines. DESIGN: The research team performed in vitro studies evaluating the anticancer efficacy via short and long-term assays and also evaluated invasion using Matrigel (Corning Life Sciences, Tewksbury, MA, USA). Further, in vitro anticancer activity of TEx was tested against 3-D cultures of HCT166 spheroids, which were subsequently analyzed by flow cytometry. SETTING: ADNA, Inc, Columbus, OH, USA; Foundation for Biomedical Research of the Academy of Athens, Athens, Greece; and Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece. INTERVENTION: The study used 4 human cell lines of colorectal cancer-HT29, HCT15, DLD1, and HCT116-and 2 breast cancer cell lines-SW480 and MDA-MB231. For a short-term assay, the extract was dissolved into culture mediums of HT29, HCT15, DLD1, HCT116, and SW480 at four 10-fold dilutions (100 to 0.1 µg/mL). For a long-term assay, TEx was added to the cultures of the same cell lines at 3 dilutions-20, 10, and 5 µg/mL. For an invasion assay, 100 µL per well of Matrigel was added and allowed to polymerize prior seeding of the MDA-MB231 cells. For cultures treated with the TEx, the TEx was mixed with the cell suspension prior to the seeding step. For the spheroid testing, the TEx was added to HCT116 cells either at the beginning of an experiment (ie, before the addition of the cancer cells), which was a chemopreventive approach, or 48 h later, on the addition of cells to the wells to allow the generation of spheroids, which was a chemotherapeutic approach. OUTCOME MEASURES: The in vitro activities of TEx were evaluated using a 48-h-incubation, short-term assay and a 2-wk, long-term (clonogenic) assay. To analyze the anti-invasive activity of the extract, images for the Matrigel invasion assay were taken with a camera at the 24-h time point. The in vitro, anticancer activity of TEx was also tested against 3-D cultures of HCT116 spheroids that were subsequently analyzed using flow cytometry. RESULTS: TEx had potently inhibited the growth of all human colon cancer cell lines tested in a dose- and time-dependent manner. TEx inhibited the formation of HCT116 spheroids when the cells were incubated with the extract. The extract also disrupted the formation of tubules formed by MDA-MB231 cells grown on Matrigel at concentrations that did not affect the overall viability of the cells, indicating a potent anti-invasive activity. CONCLUSIONS: These data suggest a potential therapeutic activity for TEx against human colon cancer, most likely due to the enhanced bioavailability of the turmeric.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Curcuma/química , Curcumina/farmacología , Extractos Vegetales/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Curcumina/química , Etanol/química , Células HCT116 , Células HT29 , Humanos , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Raíces de Plantas/química , Esferoides Celulares/efectos de los fármacos , Células Tumorales Cultivadas
2.
J Nanobiotechnology ; 9: 40, 2011 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21929751

RESUMEN

BACKGROUND: The development of effective drug delivery systems capable of transporting small interfering RNA (siRNA) has been elusive. We have previously reported that colorectal cancer tumor xenograft growth was arrested following treatment with liposomal preparation of siDCAMKL-1. In this report, we have utilized Nanoparticle (NP) technology to deliver DCAMKL-1 specific siRNA to knockdown potential key cancer regulators. In this study, mRNA/miRNA were analyzed using real-time RT-PCR and protein by western blot/immunohistochemistry. siDCAMKL-1 was encapsulated in Poly(lactide-co-glycolide)-based NPs (NP-siDCAMKL-1); Tumor xenografts were generated in nude mice, treated with NP-siDCAMKL-1 and DAPT (γ-secretase inhibitor) alone and in combination. To measure let-7a and miR-144 expression in vitro, HCT116 cells were transfected with plasmids encoding the firefly luciferase gene with let-7a and miR-144 miRNA binding sites in the 3'UTR. RESULTS: Administration of NP-siDCAMKL-1 into HCT116 xenografts resulted in tumor growth arrest, downregulation of proto-oncogene c-Myc and Notch-1 via let-7a and miR-144 miRNA-dependent mechanisms, respectively. A corresponding reduction in let-7a and miR-144 specific luciferase activity was observed in vitro. Moreover, an upregulation of EMT inhibitor miR-200a and downregulation of the EMT-associated transcription factors ZEB1, ZEB2, Snail and Slug were observed in vivo. Lastly, DAPT-mediated inhibition of Notch-1 resulted in HCT116 tumor growth arrest and down regulation of Notch-1 via a miR-144 dependent mechanism. CONCLUSIONS: These findings demonstrate that nanoparticle-based delivery of siRNAs directed at critical targets such as DCAMKL-1 may provide a novel approach to treat cancer through the regulation of endogenous miRNAs.


Asunto(s)
Carcinoma/tratamiento farmacológico , Neoplasias Colorrectales/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/farmacología , MicroARNs/metabolismo , Nanopartículas/administración & dosificación , Proteínas Serina-Treonina Quinasas/farmacología , Receptor Notch1/metabolismo , Animales , Dipéptidos/farmacología , Quinasas Similares a Doblecortina , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Nanobiotechnology ; 8: 17, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20633276

RESUMEN

BACKGROUND: The redox dye, DCPIP, has recently shown to exhibit anti-melanoma activity in vitro and in vivo. On the other hand, there is increasing evidence that synthetic nanoparticles can serve as highly efficient carriers of drugs and vaccines for treatment of various diseases. These nanoparticles have shown to serve as potent tools that can increase the bioavailability of the drug/vaccine by facilitating absorption or conferring sustained and improved release. Here, we describe results on the effects of free- and nanoparticle-enclosed DCPIP as anti-angiogenesis and anti-inflammation agents in a human colon cancer HCT116 cell line in vitro, and in induced angiogenesis in ovo. RESULTS: The studies described in this report indicate that (a) DCPIP inhibits proliferation of HCT116 cells in vitro; (b) DCPIP can selectively downregulate expression of the pro-angiogenesis growth factor, VEGF; (c) DCPIP inhibits activation of the transcriptional nuclear factor, NF-kappaB; (d) DCPIP can attenuate or completely inhibit VEGF-induced angiogenesis in the chick chorioallantoic membrane; (e) DCPIP at concentrations higher than 6 mug/ml induces apoptosis in HCT116 cells as confirmed by detection of caspase-3 and PARP degradation; and (f) DCPIP encapsulated in nanoparticles is equally or more effective than free DCPIP in exhibiting the aforementioned properties (a-e) in addition to reducing the expression of COX-2, and pro-inflammatory proteins IL-6 and IL-8. CONCLUSIONS: We propose that, DCPIP may serve as a potent tool to prevent or disrupt the processes of cell proliferation, tissue angiogenesis and inflammation by directly or indirectly targeting expression of specific cellular factors. We also propose that the activities of DCPIP may be long-lasting and/or enhanced if it is delivered enclosed in specific nanoparticles.

4.
Altern Ther Health Med ; 16(2): 12-4, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20232614

RESUMEN

Trivalent arsenic [As(III)] is currently approved by the FDA for the treatment of chronic and acute leukemias. However, As(III) has also demonstrated damaging effects on human health, including development of cardiovascular disease, diabetes, and cancer. Further, As(III) is a potent angiogenic agent. In this context, curcumin, an active ingredient in the dietary agent turmeric, has demonstrated potent antiproliferative, antiinflammatory, and antiangiogenic properties. In this report, we have shown that both curcumin and turmeric inhibit expression of vascular endothelial growth factor in HCT-116 human colon cancer cells exposed to As(III). Further, in the chicken chorioallantoic membrane assay model, treatment with low As(III) concentrations results in extensive increase in blood vessel density, which, however, is reduced in the presence of curcumin or turmeric. Collectively, the findings reported here strongly suggest that turmeric and curcumin can dramatically attenuate the process of angiogenesis induced by low As(III) concentrations.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Arsénico/efectos adversos , Neoplasias del Colon/tratamiento farmacológico , Curcuma/química , Curcumina/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Arsénico/uso terapéutico , Embrión de Pollo , Humanos , Leucemia/tratamiento farmacológico , Fitoterapia
6.
Methods Mol Biol ; 906: 311-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22791444

RESUMEN

Nanoparticles (NPs) formulated using poly (D,L-lactide-co-glycolide) (PLGA), a biodegradable, biocompatible, and clinically approved polymer, have been widely used for targeted drug delivery. Here we provide methods for preparing PLGA NPs that encapsulate small interfering RNA (siRNA). The siRNA NPs are formulated using a double-emulsion solvent evaporation technique with the addition of a small amount of the cationic polymer, polyethyleneimine, which significantly increases siRNA encapsulation.


Asunto(s)
Ácido Láctico/química , Nanopartículas/química , Ácido Poliglicólico/química , ARN Interferente Pequeño/administración & dosificación , Sistemas de Liberación de Medicamentos , Emulsiones/química , Nanopartículas/ultraestructura , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Interferente Pequeño/química , Solventes/química
7.
In Vivo ; 24(6): 861-4, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21164045

RESUMEN

BACKGROUND: curcumin metabolites are detectable in body fluids such as serum and urine. We have developed a novel assay that can detect metabolites in such body fluids by measuring their effect on the nuclear factor kappa B/inhibitor of kappa B (NF-κB/IκB) pathway. PATIENTS AND METHODS: fifteen healthy individuals were enrolled in the study and randomly assigned to two groups: control group (five) and curcumin group (ten). The test group ingested 8 g of the curcuminoids (C(3)-Complex) with 16 oz of bottled water. Blood and urine were collected at 0, 4, 8, and 24 h after ingestion. Degradation of the NF-κB/IκB complex was detected by the Genetic Expression and Measurement (GEM) assay using HCT116 cells stably transfected with PGL3-IκB firefly luciferase. RESULTS: using our novel GEM assay, the five controls who had not taken curcumin were identified. CONCLUSION: the GEM assay is a very sensitive and accurate non-invasive assay that could be utilized to detect metabolites in body fluids. It could also serve as a tool to determine participants' compliance during clinical research studies.


Asunto(s)
Curcumina/farmacocinética , Proteínas I-kappa B/metabolismo , Biomarcadores/sangre , Biomarcadores/orina , Estudios de Factibilidad , Femenino , Células HCT116 , Humanos , Luciferasas de Luciérnaga/genética , Masculino , FN-kappa B/metabolismo , Proyectos Piloto , Sensibilidad y Especificidad
8.
Cancer Res ; 68(6): 1962-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18339878

RESUMEN

Diphenyl difluoroketone (EF24), a molecule having structural similarity to curcumin, was reported to inhibit proliferation of a variety of cancer cells in vitro. However, the efficacy and in vivo mechanism of action of EF24 in gastrointestinal cancer cells have not been investigated. Here, we assessed the in vivo therapeutic effects of EF24 on colon cancer cells. Using hexosaminidase assay, we determined that EF24 inhibits proliferation of HCT-116 and HT-29 colon and AGS gastric adenocarcinoma cells but not of mouse embryo fibroblasts. Furthermore, the cancer cells showed increased levels of activated caspase-3 and increased Bax to Bcl-2 and Bax to Bcl-xL ratios, suggesting that the cells were undergoing apoptosis. At the same time, cell cycle analysis showed that there was an increased number of cells in the G(2)-M phase. To determine the effects of EF24 in vivo, HCT-116 colon cancer xenografts were established in nude mice and EF24 was given i.p. EF24 significantly suppressed the growth of colon cancer tumor xenografts. Immunostaining for CD31 showed that there was a lower number of microvessels in the EF24-treated animals coupled with decreased cyclooxygenase-2, interleukin-8, and vascular endothelial growth factor mRNA and protein expression. Western blot analyses also showed decreased AKT and extracellular signal-regulated kinase activation in the tumors. Taken together, these data suggest that the novel curcumin-related compound EF24 is a potent antitumor agent that induces caspase-mediated apoptosis during mitosis and has significant therapeutic potential for gastrointestinal cancers.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Compuestos de Bencilideno/farmacología , Neoplasias del Colon/tratamiento farmacológico , Piperidonas/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Células HCT116 , Células HT29 , Humanos , Interleucina-8/biosíntesis , Interleucina-8/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res ; 68(20): 8573-81, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922933

RESUMEN

We have identified a natural compound that activates apoptosis of epithelial cancer cells through activation of tumor necrosis factor-alpha (TNF-alpha), TNF receptor (TNFR)-associated death domain (TRADD), and caspases. The molecule 1-hydroxy-5,7-dimethoxy-2-naphthalene-carboxaldehyde (HDNC, marmelin) was isolated and characterized from ethyl acetate fraction of extracts of Aegle marmelos. HDNC treatment inhibited the growth of HCT-116 colon cancer tumor xenografts in vivo. Immunostaining for CD31 showed that there was a significant reduction in microvessels in the HDNC-treated animals, coupled with decreased cyclooxygenase-2, interleukin-8, and vascular endothelial growth factor mRNA. Using hexoseaminidase assay, we determined that HDNC inhibits proliferation of HCT-116 colon and HEp-2 alveolar epithelial carcinoma cells. Furthermore, the cancer cells showed increased levels of activated caspase-3 and induced G(1) cell cycle arrest, which was suppressed by caspase-3 inhibitors. HDNC induced TNF-alpha, TNFR1, and TRADD mRNA and protein expression. Moreover, caspase-8 and Bid activation, and cytochrome c release, were observed, suggesting the existence of a cross-talk between death receptor and the mitochondrial pathways. HDNC inhibited AKT and extracellular signal-regulated kinase phosphorylation both in cells in culture and in tumor xenografts. In addition, electrophoretic mobility shift assay and luciferase reporter assays showed that HDNC significantly suppressed TNF-alpha-mediated activation and translocation of nuclear factor-kappaB (NF-kappaB). This was further confirmed by Western blot analysis of nuclear extracts wherein levels of RelA, the p65 component of NF-kappaB, were significantly less in cells treated with HDNC. Together, the data suggest that the novel compound HDNC (marmelin) is a potent anticancer agent that induces apoptosis during G(1) phase of the cell cycle and could be a potential chemotherapeutic candidate.


Asunto(s)
Aegle/química , Antineoplásicos Fitogénicos/farmacología , Naftoles/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fase G1/efectos de los fármacos , Células HCT116 , Humanos , Ratones , FN-kappa B/antagonistas & inhibidores , Neoplasias Experimentales/tratamiento farmacológico , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/análisis , Proteína de Dominio de Muerte Asociada a Receptor de TNF/genética , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA